1
|
Harada K, Sakamoto N, Kitaoka T, Nakamura Y, Kondo R, Morisue R, Hashimoto H, Yamamoto Y, Ukai S, Maruyama R, Sakashita S, Kojima M, Tanabe K, Ohdan H, Shitara K, Kinoshita T, Ishii G, Yasui W, Ochiai A, Ishikawa S. PI3 expression predicts recurrence after chemotherapy with DNA-damaging drugs in gastric cancer. J Pathol 2025; 265:472-485. [PMID: 39980125 PMCID: PMC11880974 DOI: 10.1002/path.6400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/10/2024] [Accepted: 01/06/2025] [Indexed: 02/22/2025]
Abstract
Despite recent advances in gastric cancer therapy, chemotherapy resistance and lack of methods for selecting combination regimens remain major problems. Organoids, which provide a culture system that more closely resembles tumor cell organization than traditional cell lines, can be established from surgical specimens with a high success rate and are widely used for drug sensitivity assays. In this study, we aimed to identify a novel biomarker for predicting multidrug resistance using gastric cancer organoids (GCOs). We evaluated 5-fluorouracil or oxaliplatin-resistant GCOs to find novel biomarkers that reflect multidrug resistance in gastric cancer. To examine the resistance mechanisms, RNA-sequencing analysis and ex vivo drug sensitivity testing were performed. The association of biomarkers with patient prognosis and chemotherapy efficacy was evaluated using three original cohorts with a total of 230 cases. The results were also validated with two independent public cohorts and single-cell RNA sequence data. Increased expression of peptidase inhibitor 3 (PI3) was detected in all 5-fluorouracil or oxaliplatin-resistant GCOs. Our findings suggest a potential association of PI3 expression with ribosome biosynthesis and RNA metabolism under organoid conditions. We also found that PI3 overexpression promoted 5-fluorouracil/oxaliplatin/cisplatin resistance but not paclitaxel resistance. Immunohistochemical evaluation of PI3 expression revealed that the PI3-positive gastric cancer group had a poorer outcome, especially in terms of time to recurrence. PI3 positivity was also an independent predictor of relapse after chemotherapy with DNA-damaging agents. PI3 promotes DNA-damaging drug resistance through multiple downstream regulations related to RNA and ribosomal metabolism. PI3 may be useful as a biomarker for the therapeutic selection of non-DNA-damaging agents. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kenji Harada
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Naoya Sakamoto
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
| | - Takumi Kitaoka
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
- The Department of Pathology, Faculty of MedicineYamagata UniversityYamagataJapan
| | - Yuka Nakamura
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Ryotaro Kondo
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Ryo Morisue
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Hepatobiliary and Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Hiroko Hashimoto
- Division of Innovative Pathology and Laboratory MedicineExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Yusuke Yamamoto
- Division of Molecular and Cellular MedicineNational Cancer Center Research InstituteTokyoJapan
| | - Shoichi Ukai
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Ryota Maruyama
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Shingo Sakashita
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
| | - Motohiro Kojima
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
| | - Kazuaki Tanabe
- Department of Perioperative and Critical Care Management, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Takahiro Kinoshita
- Division of Gastric SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Genichiro Ishii
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
- Division of Innovative Pathology and Laboratory MedicineExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Wataru Yasui
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Atsushi Ochiai
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Shumpei Ishikawa
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Preventive Medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
2
|
Laplante P, Rosa R, Nebot-Bral L, Goulas J, Pouvelle C, Nikolaev S, Silvin A, Kannouche PL. Effect of MisMatch repair deficiency on metastasis occurrence in a syngeneic mouse model. Neoplasia 2025; 62:101145. [PMID: 39985912 PMCID: PMC11905862 DOI: 10.1016/j.neo.2025.101145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Mismatch repair deficiency leads to high mutation rates and microsatellite instability (MSI-H), associated with immune infiltration and responsiveness to immunotherapies. In early stages, MSI-H tumors generally have a better prognosis and lower metastatic potential than microsatellite-stable (MSS) tumors, especially in colorectal cancer. However, in advanced stages, MSI-H tumors lose this survival advantage for reasons that remain unclear. We developed a syngeneic mouse model of MSI cancer by knocking out the MMR gene Msh2 in the metastatic 4T1 breast cancer cell line. This model mirrored genomic features of MSI-H cancers and showed reduction in metastatic incidence compared to their MSS counterparts. In MSI-H tumors, we observed an enrichment of immune gene-signatures that negatively correlated with metastasis incidence. A hybrid epithelial-mesenchymal signature, related to aggressiveness was detected only in metastatic MSI-H tumors. Interestingly, we identified immature myeloid cells at primary and metastatic sites in MSI-H tumor-bearing mice, suggesting that MMR deficiency elicits specific immune responses beyond T-cell activation.
Collapse
Affiliation(s)
- Pierre Laplante
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Reginaldo Rosa
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Laetitia Nebot-Bral
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Jordane Goulas
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Caroline Pouvelle
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Sergey Nikolaev
- Paris-Saclay Université, Inserm-U981, Gustave Roussy, Villejuif, France
| | - Aymeric Silvin
- Paris-Saclay Université, Inserm-U1015, Gustave Roussy, Villejuif, France
| | - Patricia L Kannouche
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France.
| |
Collapse
|
3
|
Heslin RT, Whitham ZA, Kim AC. Molecular and Genetic Markers of Peritoneal Metastasis. Surg Oncol Clin N Am 2025; 34:145-154. [PMID: 40015796 DOI: 10.1016/j.soc.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Peritoneal surface malignancies (PSMs) represent a biologically diverse group of cancers that range from primary peritoneal mesothelioma to metastatic gastrointestinal cancers. Because of the heterogenous nature of PSM, there is a large gap in molecular characterization of these cancers. This article reviews the underlying molecular and genetic mechanisms for PSM.
Collapse
Affiliation(s)
- Ryan T Heslin
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Zachary A Whitham
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Alex C Kim
- Department of Surgical Oncology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Lim CY, Cha DI, Jeong WK, Cho YY, Hong S, Hong S, Kim K, Kim JH. Prediction of microsatellite-stable/epithelial-to-mesenchymal transition molecular subtype gastric cancer using CT radiomics and clinicopathologic factors. Eur J Radiol 2025; 185:111990. [PMID: 39956084 DOI: 10.1016/j.ejrad.2025.111990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/07/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
OBJECTIVES This study aimed to develop a predictive model for the microsatellite-stable (MSS)/epithelial-to-mesenchymal transition (EMT) subtype of gastric cancer (GC) using computed tomography (CT) radiomics and clinicopathological factors. MATERIALS AND METHODS This retrospective study included 418 patients with GC who underwent primary resection and transcriptome analysis with microarray between October 1995 and May 2008. Using preoperative CT images, radiomic features from the volume of interest in the portal venous phase images were extracted. The patient data were randomly divided into training (70%) and testing (30%) datasets. Optimal radiomics features were selected through a thorough feature-selection process. The final radiomic and clinicopathological factors were selected using a stepwise variable selection method. The area under the curve (AUC) was calculated to evaluate performance. RESULTS Seventy patients had EMT subtype GC, and 348 patients had non-EMT subtype based on transcriptome analysis. There were 276 men (66.0 %), with a median age of 59 years (interquartile range: 50-67). Eleven radiomic features were selected for the prediction model using the combined variance inflation factor (VIF) and least absolute shrinkage and selection operator (LASSO) method. A CT radiomics-based prediction model was constructed using logistic regression with AUCs of 0.824 and 0.736 for training and testing, respectively. When clinicopathological factors such as age, tumor size, signet ring cell histology, and Lauren classification were combined, the AUCs of the models increased to 0.849 and 0.840 for training and testing, respectively (p < 0.001 for testing). CONCLUSION A prediction model using CT radiomics and clinicopathological factors showed good performance in predicting the EMT subtype of GC.
Collapse
Affiliation(s)
- Chae Young Lim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Ik Cha
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Woo Kyoung Jeong
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon Young Cho
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Sungjun Hong
- Department of Digital Health, Samsung Advanced Institute of Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea; Medical AI Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Sungsoo Hong
- Department of Digital Health, Samsung Advanced Institute of Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Kyunga Kim
- Department of Digital Health, Samsung Advanced Institute of Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea; Biomedical Statistics Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea; Department of Data Convergence & Future Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae-Hun Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
5
|
Cho SY, Hwang H, Lee HS, Kwon Y, Khanh Vu N, Baek JG, Jeon M, Bae J, Kwon HC, Kim WK, Kwon J. Chemical constituents from the Korean endemic plant Pseudolysimachion pusanensis inhibit diffuse-type gastric cancer cells. Biomed Pharmacother 2025; 186:118005. [PMID: 40138921 DOI: 10.1016/j.biopha.2025.118005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Diffuse-type gastric cancer (GC) is closely associated with genetic abnormalities; however, its exact pathological mechanisms are still not understood. It manifests without symptoms before advanced stages and is often difficult to diagnose using routine imaging tests. Therefore, specific targeted therapies for diffuse GC are currently unavailable. In this study, the extract of Pseudolysimachion pusanensis, which is endemic to Korea, inhibited the proliferation of GC cells, MKN1 and SNU668, while the other extracts of the two endemic Pseudolysimachion species did not show any activity. This led to the molecular networking analysis of Pseudolysimachion species to identify the molecules mostly observed only in P. pusanensis. Thirteen new (1-13) and eight known (14-21) compounds were obtained and structurally characterized. Of these, cucurbitacin derivative compounds 1 and 14-16 showed activity, and particularly, the IC50 value of compound 1 was 0.65 and 0.21 μM. Ki-67 expression analysis, single-cell originated cell proliferation assay, and western blot analysis of apoptotic cell death-related molecules revealed that compound 1 mediated both cytostasis and cellular death via apoptosis. Particularly, this compound exhibited an anti-tumorigenic effect on the invasion of diffuse-type GC cells by perturbing the epithelial-to-mesenchymal transition (EMT) pathway, as demonstrated by invasion assays using both 2D models and in vivo-like 3D spheroid co-culture models, as well as western blot analysis of EMT markers. In addition, some functional groups that may or may not be necessary for the activity of cucurbitacin derivatives were identified, and some clues that the presence of metal ions may affect the activity were provided.
Collapse
Affiliation(s)
- Su-Yeon Cho
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hoseong Hwang
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Hyeon-Seong Lee
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Yujin Kwon
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Ngoc Khanh Vu
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Jong Gwon Baek
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Mukyeong Jeon
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Joonbeom Bae
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hak Cheol Kwon
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Won Kyu Kim
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea.
| | - Jaeyoung Kwon
- KIST Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea; Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea; Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea.
| |
Collapse
|
6
|
Zhu W, Sun J, Jing F, Xing Y, Luan M, Feng Z, Ma X, Wang Y, Jia Y. GLI2 inhibits cisplatin sensitivity in gastric cancer through DEC1/ZEB1 mediated EMT. Cell Death Dis 2025; 16:204. [PMID: 40133270 PMCID: PMC11937514 DOI: 10.1038/s41419-025-07564-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Cisplatin (CDDP) based chemotherapy has emerged as the predominant therapeutic regimen for patients with advanced gastric cancer (GC). However, its efficacy is dampened by the development of chemoresistance, which results in poor prognosis of patients. GLI2, a key transcription factor in the Hedgehog (Hh) signaling pathway, is regarded as a target for cancer therapy. However, the significance of GLI2 for CDDP resistance in GC has not been well established. Here, we show that GLI2 expression was upregulated in EMT-type GC and associated with poor prognosis. GLI2 promotes proliferation, migration, and CDDP resistance of GC cells by inducing EMT. In terms of mechanism, GLI2 binds to the promoter region of DEC1 and enhances its expression, thereby co-transcriptionally regulating ZEB1 expression. Animal experiments have demonstrated that both GLI2 knockdown and GLI2 inhibitor significantly enhance CDDP sensitivity in GC. Our data not only identify a novel GLI2/DEC1/ZEB1/EMT pathway in GC CDDP resistance but also provide novel strategies to treat GC in the future.
Collapse
Affiliation(s)
- Wenshuai Zhu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Jingguo Sun
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Fubo Jing
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China
| | - Muhua Luan
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China
| | - Zhaotian Feng
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, People's Republic of China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China
| | - Yunshan Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China.
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China.
| | - Yanfei Jia
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China.
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, People's Republic of China.
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, People's Republic of China.
| |
Collapse
|
7
|
Li R, Yang T, Dong Z, Gao Y, Li N, Song T, Sun J, Chen Y. Factors influencing the incidence of early gastric cancer: a bayesian network analysis. BMC Gastroenterol 2025; 25:194. [PMID: 40119277 PMCID: PMC11927266 DOI: 10.1186/s12876-025-03765-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/06/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND This study aims to establish a Bayesian network risk prediction model for gastric cancer using data mining methods. It explores both direct and indirect factors influencing the incidence of gastric cancer and reveals the interrelationships among these factors. METHODS Data were collected from early cancer screenings conducted at the People's Hospital of Lincang between 2022 and 2023. Initial variable selection was performed using Least Absolute Shrinkage and Selection Operator (Lasso) and Sliding Windows Sequential Forward Selection (SWSFS), and the screened variables and demographic characteristics features were used as variables for constructing the Bayesian network (BN) model. Subsequently, the performance of the models was evaluated, and the optimal model was selected for network mapping and Bayesian inference using the best model. RESULTS The incidence rate of gastric cancer in this region's high-risk population was determined to be 7.09%. The BN model constructed from the set of variables consisting of Lasso's selection variables and demographic characteristics had better performance. A total of 12 variables were incorporated into the BN model to form a network structure consisting of 13 nodes and 18 edges. The model shows that age, gender, ethnicity, current address, upper gastrointestinal symptoms (nausea, acid reflux, vomiting), alcohol consumption, smoking, SGIM gastritis, and family history are important risk factors for gastric cancer development. CONCLUSION The Bayesian network model provides an intuitive framework for understanding the direct and indirect factors contributing to the early onset of gastric cancer, elucidating the interrelationships among these factors. Furthermore, the model demonstrates satisfactory predictive performance, which may facilitate the early detection of gastric cancer and enhance the levels of early diagnosis and treatment among high-risk populations.
Collapse
Affiliation(s)
- Ruiyu Li
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Taiming Yang
- Department of Gastroenterology, The People's Hospital of Lincang, Lincang, 677000, Yunnan, China
| | - Zi Dong
- Department of Gastroenterology, The People's Hospital of Lincang, Lincang, 677000, Yunnan, China
| | - Yin Gao
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Nan Li
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Ting Song
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jinshu Sun
- Department of Gastroenterology, The People's Hospital of Lincang, Lincang, 677000, Yunnan, China
| | - Ying Chen
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
8
|
Bounder G, Jouimyi MR, Essaidi I, Elyounsi I, Boura H, Michel V, Badre W, Touati E, Maachi F. Upstream stimulating factor 1 (USF1) -202 G/A polymorphism and serum levels of USF1 and USF2 are associated with gastric cancer risk: a case control study. J Cancer Res Clin Oncol 2025; 151:113. [PMID: 40102295 PMCID: PMC11919976 DOI: 10.1007/s00432-025-06158-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE Gastric cancer is an inflammation-driven disease often associated with a bad prognosis. Upstream stimulatory factors USF1 and USF2 are pleiotropic transcription factors, with tumor suppressor function. Low expression of USF1 is associated with low survival in gastric cancer patients. USF1 genetic polymorphism -202G > A has been associated with cancer susceptibility. Our aim was to investigate USF1 gene polymorphism and serum level with the risk of gastric cancer. METHODS USF1-202 G/A polymorphism was analyzed by sanger sequencing, with the measure of USF1/USF2 serum levels by ELISA in H. pylori-positive patients with chronic gastritis, gastric precancerous lesions, gastric cancer and in healthy controls. RESULTS Our results show that the presence of the USF1-202 A allele increased the risk of gastric cancer compared to G (OR = 2; 95% CI 1.07-3.9; P = 0.02). Genotypically and under the dominant mutation model, the combined USF1- GA/AA -202 genotypes corresponded to higher risk of gastric cancer (OR = 3.5; 95% CI 1.4-8.2; p-value = 0.005) than the GG genotype. Moreover, the G/A transition at USF1-202 was associated with lower USF1 serum level, and mostly observed in gastric cancer patients where the average serological level of USF1 were 2.3 and twofold lower for the AA and GA genotypes, respectively, compared to GG. CONCLUSION USF1-202 G/A polymorphism constitutes a gastric cancer genetic risk factor. Together with USF1/USF2 serum level, they can be proposed as promising biomarkers for gastric cancer detection/prevention.
Collapse
Grants
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
- ACIP2015-10 INSTITUT PASTEUR Paris, as a Pasteur International Concerted action,
Collapse
Affiliation(s)
- Ghizlane Bounder
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Mohamed Reda Jouimyi
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Imane Essaidi
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | | | - Hasna Boura
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Valérie Michel
- Équipe DMic01-Infection, Génotoxicité et Cancer, Département de Microbiologie, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Institut Pasteur, Université Paris Cité, 75015, Paris, France
| | - Wafa Badre
- Gastroenterology Department, Ibn Rochd University Hospital Center, Casablanca, Morocco
| | - Eliette Touati
- Équipe DMic01-Infection, Génotoxicité et Cancer, Département de Microbiologie, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6047, Institut Pasteur, Université Paris Cité, 75015, Paris, France.
| | - Fatima Maachi
- Helicobacter Pylori and Gastric Pathologies Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco.
| |
Collapse
|
9
|
Veas Rodriguez J, Piñol M, Sorolla MA, Parisi E, Sorolla A, Santacana M, Ruiz M, Parra G, Bernabeu M, Iglesias M, Aracil C, Escartin A, Vilardell F, Matias-Guiu X, Salud A, Montal R. Comprehensive immunophenotyping of gastric adenocarcinoma identifies an inflamed class of tumors amenable to immunotherapies. J Immunother Cancer 2025; 13:e010024. [PMID: 40102027 PMCID: PMC11927434 DOI: 10.1136/jitc-2024-010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Gastric adenocarcinoma (GAC) imposes a considerable global health burden. Molecular profiling of GAC from the tumor microenvironment perspective through a multi-omics approach is eagerly awaited in order to allow a more precise application of novel therapies in the near future. METHODS To better understand the tumor-immune interface of GAC, we identified an internal cohort of 82 patients that allowed an integrative molecular analysis including mutational profiling by whole-exome sequencing, RNA gene expression of 770 genes associated with immune response, and multiplex protein expression at spatial resolution of 34 immuno-oncology targets at different compartments (tumorous cells and immune cells). Molecular findings were validated in 595 GAC from the TCGA and ACRG external cohorts with available multiomics data. Prediction of response to immunotherapies of the discovered immunophenotypes was assessed in 1039 patients with cancer from external cohorts with available transcriptome data. RESULTS Unsupervised clustering by gene expression identified a subgroup of GAC that includes 52% of the tumors, the so-called Inflamed class, characterized by high tumor immunogenicity and cytotoxicity, particularly in the tumor center at protein level, with enrichment of PIK3CA and ARID1A mutations and increased presence of exhausted CD8+ T cells as well as co-inhibitory receptors such as PD1, CTLA4, LAG3, and TIGIT. The remaining 48% of tumors were called non-inflamed based on the observed exclusion of T cell infiltration, with an overexpression of VEGFA and higher presence of TP53 mutations, resulting in a worse clinical outcome. A 10-gene RNA signature was developed for the identification of tumors belonging to these classes, demonstrating in evaluated datasets comparable clinical utility in predicting response to current immunotherapies when tested against other published gene signatures. CONCLUSIONS Comprehensive immunophenotyping of GAC identifies an inflamed class of tumors that complements previously proposed tumor-based molecular clusters. Such findings may provide the rationale for exploring novel immunotherapeutic approaches for biomarker-enriched populations in order to improve GAC patient's survival.
Collapse
Affiliation(s)
- Joel Veas Rodriguez
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Miquel Piñol
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Alba Sorolla
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Eva Parisi
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Anabel Sorolla
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Santacana
- Scientific and Technical Service of Immunohistochemistry, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Ruiz
- Scientific and Technical Service of Biobank, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Genís Parra
- CNAG-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mario Bernabeu
- CNAG-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mar Iglesias
- Department of Pathology, Hospital del Mar, University Pompeu Fabra, Hospital del Mar Research Institute, CIBERONC, Barcelona, Spain
| | - Carles Aracil
- Department of Gastroenterology, Clinical and Experimental Research in Digestive and Hematological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Alfredo Escartin
- Department of Surgery, Experimental Surgery Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Felip Vilardell
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Antonieta Salud
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Robert Montal
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| |
Collapse
|
10
|
Yuan Z, Wang JH, Cui H, Wang SY, Wei B, Cui JX. Mapping the landscape of gastric cancer immunotherapy: Bibliometric insights into advances and hotspots. World J Gastrointest Oncol 2025; 17:100997. [PMID: 40092931 PMCID: PMC11866247 DOI: 10.4251/wjgo.v17.i3.100997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/11/2024] [Accepted: 12/31/2024] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Immunotherapy has surfaced as a promising therapeutic modality for gastric cancer (GC). A comprehensive review of advancements, current status, and research trends in GC immunotherapy is essential to inform future investigative efforts. AIM To delineate the trends, advancements, and focal points in immunotherapy for GC. METHODS We performed a bibliometric analysis of 2906 articles in English concerning GC immunotherapy published from 2000 to December 20, 2023, indexed in the Web of Science Core Collection. Data analysis and visualization were facilitated by CiteSpace (6.1.6R), VOSviewer v.1.6.17, and GraphPad Prism v8.0.2. RESULTS There has been an increase in the annual publication rate of GC immunotherapy research. China leads in publication volume, while the United States demonstrates the highest citation impact. Fudan University is notable for its citation frequency and publication output. Co-citation analysis and keyword frequency revealed and highlighted a focus on GC prognosis, the tumor microenvironment (TME), and integrative immunotherapy with targeted therapy. Emerging research areas include gastroesophageal junction cancer, adoptive immunotherapy, and the role of Treg cell in immunotherapy. CONCLUSION GC immunotherapy research is an expanding field attracting considerable scientific interest. With the clinical adoption of immunotherapy in GC, the primary goals are to enhance treatment efficacy and patient outcomes. Unlike hematological malignancies, GC's solid TME presents distinct immunological challenges that may attenuate the cytotoxic effects of immune cells on cancer cells. For instance, although CAR-T therapy is effective in hematological malignancies, it has underperformed in GC settings. Current research is centered on overcoming immunosuppression within the TME, with a focus on combinations of targeted therapy, adoptive immunotherapy, Treg cell dynamics, and precise prognosis prediction in immunotherapy. Additionally, immunotherapy's role in treating gastroesophageal junction cancer has become a novel research focus.
Collapse
Affiliation(s)
- Zhen Yuan
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing-Hang Wang
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Hao Cui
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Shu-Yuan Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Bo Wei
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jian-Xin Cui
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
11
|
Xu R, He D, Sun R, Zhou J, Xin M, Liu Q, Dai Y, Li H, Zhang Y, Li J, Shan X, He Y, Xu B, Guo Q, Ning S, Gao Y, Wang P. CNV-mediated dysregulation of the ceRNA network mechanism revealed heterogeneity in diffuse and intestinal gastric cancers. J Transl Med 2025; 23:308. [PMID: 40069783 PMCID: PMC11895245 DOI: 10.1186/s12967-025-06222-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/11/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a highly heterogeneous tumour with high morbidity. Approximately 95% of GC cases are gastric adenocarcinomas, which are further categorized into two predominant subtypes: diffuse gastric cancer (DGC) and intestinal gastric cancer (IGC). These subtypes exhibit distinct pathophysiological and molecular characteristics, reflecting their unique tumorigenic mechanisms. METHOD In this study, we employed a comprehensive approach to identify driver genes associated with DGC and IGC by focusing on copy number variation (CNV) genes within the competing endogenous RNA (ceRNA) network. The influence of driver CNV genes on the molecular, cellular, and clinical differences between DGC and IGC was subsequently analysed. Finally, therapeutic strategies for DGC and IGC were evaluated based on the status and functional pathways of the driver CNV genes. RESULTS A total of 17 and 22 driver CNV genes were identified in DGC and IGC, respectively. These genes drive subtype differences through the ceRNA network, resulting in alterations in the tumour microenvironment (TME). Based on these differences, personalized treatment strategies for DGC or IGC could be developed. Immune checkpoint inhibitors may be an effective treatment option in IGC. Additionally, DGC patients with homozygous deletion of PPIF might benefit from adjuvant chemotherapy, whereas those with high-level amplification of MTAP could respond to targeted therapy. CONCLUSION Driver CNV genes were identified to reveal the underlying cause of heterogeneity in DGC and IGC. Furthermore, specific driver CNV genes were identified as potential therapeutic targets, facilitating personalized treatment.
Collapse
Affiliation(s)
- Rongji Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Danni He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Rui Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Jiaqi Zhou
- The First Clinical School of Gansu University of Chinese Medicine, Lanzhou, 730030, China
| | - Mengyu Xin
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Qian Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yifan Dai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Houxing Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yujie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Jiatong Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - XinXin Shan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yuting He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Borui Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Qiuyan Guo
- The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| | - Yue Gao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| | - Peng Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
12
|
Cammarota A, Woodford R, Smyth EC. Targeting HER2 in Gastroesophageal Cancer: A New Appetite for an Old Plight. Drugs 2025; 85:361-383. [PMID: 39843758 DOI: 10.1007/s40265-024-02132-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/24/2025]
Abstract
The incidence of gastroesophageal cancers is rising, driven, in part, by an increasing burden of risk factors of obesity and gastroesophageal reflux. Despite efforts to address these risk factors, and a growing interest in methods of population screening, the bulk of these tumours are unresectable at diagnosis. In this setting, effective systemic treatments are paramount to improve survival and quality of life. Early and accurate identification of oncogenic drivers, such as human epidermal growth factor receptor 2 (HER2), present in 5-30% of gastroesophageal adenocarcinomas (GEAs), is integral to guide choice of therapies due to the clear predictive implications that arise from overexpression of this receptor. After trastuzumab, the first anti-HER2 agent with approved use in HER2-positive GEA, the addition of pembrolizumab to first-line trastuzumab-chemotherapy and trastuzumab deruxtecan in the refractory space have more recently changed practice. Yet, the response to these agents has been vastly different across patients with HER2-positive disease, underpinning the need for reliable biomarkers of response. Emergent data have suggested that levels of HER2 expression on tissue or liquid biopsies may predict response to first-generation HER2 therapies while HER2 heterogeneity, receptor changes, co-occurring molecular alterations and oncogenic genomic and metabolic reprogramming may be implicated in resistance. A robust knowledge of the mechanisms of resistance and response to HER2-directed therapies is necessary to inform novel strategies of HER2-targeting and guide choice combinations with other biomarker-directed therapies, to improve outcomes from a new generation of clinical trials in HER2-positive GEA. Understanding and close examination of previous failures in this space form an important part of this assessment, as does correlative biomarker and translational work pertaining to the role of HER2 and dynamic changes that result through treatment exposure. In this review, we aim to provide an overview of strategies for HER2 targeting, summarising both the successes and disappointments in this therapeutic landscape and discuss existing challenges and future perspectives on development in this highly morbid tumour type.
Collapse
Affiliation(s)
- Antonella Cammarota
- Sarah Cannon Research Institute UK, 93 Harley St, London, UK
- Department of Medical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan, Italy
| | - Rachel Woodford
- Sarah Cannon Research Institute UK, 93 Harley St, London, UK
- National Health and Medical Research Council Clinical Trials Centre (NHMRC CTC), University of Sydney, Parramatta Road, Camperdown, Australia
| | - Elizabeth C Smyth
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
13
|
Yang Z, Ren C, He Z, Luo B, Chen X, Xu E, Guan W, Xia X. Identification of AXL as a novel positive regulator of lipid raft in gastric cancer. Cell Signal 2025; 127:111573. [PMID: 39708896 DOI: 10.1016/j.cellsig.2024.111573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/30/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Lipid rafts are highly heterogeneous and dynamic microdomains involved in molecule trafficking and signaling transduction. This study investigates the role of lipid rafts in gastric cancer and their key regulators. Analyzing FFPE samples from 111 gastric cancer patients, we found that high lipid raft levels predict poor prognosis. Modulating these levels in gastric cancer cell lines significantly impacted cell proliferation, migration, and invasion. Weighted Gene Co-expression Network Analysis identified AXL as a hub gene associated with lipid rafts. AXL knockdown experiments revealed its interaction with Caveolin-1, a caveolae lipid raft protein, which regulates lipid raft levels and promotes AKT and ERK signaling, enhancing cancer development and metastasis. In vivo tumorigenesis assays and survival analyses further supported these findings. This study underscores the significance of lipid rafts in gastric cancer and identifies AXL as a novel regulator, offering new insights into the molecular mechanisms of cancer progression and suggesting potential therapeutic targets.
Collapse
Affiliation(s)
- Zhi Yang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chuanfu Ren
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Ziyun He
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Banxin Luo
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Chen
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - En Xu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Wenxian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China; Department of General Surgery, Taikang Xianlin DrumTower Hospital, Nanjing, China.
| | - Xuefeng Xia
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China; Department of General Surgery, Taikang Xianlin DrumTower Hospital, Nanjing, China.
| |
Collapse
|
14
|
Yang B, Li W, Xu Z, Li W, Hu G. NetSDR: Drug repurposing for cancers based on subtype-specific network modularization and perturbation analysis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167688. [PMID: 39862994 DOI: 10.1016/j.bbadis.2025.167688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/04/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Cancer, a heterogeneous disease, presents significant challenges for drug development due to its complex etiology. Drug repurposing, particularly through network medicine approaches, offers a promising avenue for cancer treatment by analyzing how drugs influence cellular networks on a systemic scale. The advent of large-scale proteomics data provides new opportunities to elucidate regulatory mechanisms specific to cancer subtypes. Herein, we present NetSDR, a Network-based Subtype-specific Drug Repurposing framework for prioritizing repurposed drugs specific to certain cancer subtypes, guided by subtype-specific proteomic signatures and network perturbations. First, by integrating cancer subtype information into a network-based method, we developed a pipeline to recognize subtype-specific functional modules. Next, we conducted drug response analysis for each module to identify the "therapeutic module" and then used deep learning to construct weighted drug response network for the particular subtype. Finally, we employed a perturbation response scanning-based drug repurposing method, which incorporates dynamic information, to facilitate the prioritization of candidate drugs. Applying the framework to gastric cancer, we attested the significance of the extracellular matrix module in treatment strategies and discovered a promising potential drug target, LAMB2, as well as a series of possible repurposed drugs. This study demonstrates a systems biology framework for precise drug repurposing in cancer and other complex diseases.
Collapse
Affiliation(s)
- Bin Yang
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Wanshi Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Zhen Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Guang Hu
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; Key Laboratory of Alkene-carbon Fibres-based Technology & Application for Detection of Major Infectious Diseases, Soochow University, Suzhou 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China.
| |
Collapse
|
15
|
Komori A, Hironaka S, Kadowaki S, Mitani S, Furuta M, Kawakami T, Makiyama A, Takegawa N, Sugiyama K, Hirano H, Ando T, Matsushima T, Chida A, Kashiwada T, Komoda M, Matsumoto T, Oda H, Yabusaki H, Kawakami H, Yamazaki K, Boku N, Hyodo I, Yoshimura K, Muro K. Prevalence and clinicopathological features of microsatellite instability-high metastatic or recurrent gastric and esophagogastric junction cancer: WJOG13320GPS. Gastric Cancer 2025; 28:301-308. [PMID: 39738793 DOI: 10.1007/s10120-024-01579-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Microsatellite instability (MSI)-high tumors represent a distinct, small-fraction subtype in esophagogastric junction cancer or gastric cancer (GC), yet their clinical significance remains poorly understood. This study aimed to investigate the prevalence and clinicopathological features of chemotherapy-naïve metastatic or recurrent MSI-high GC as a prescreening study for a phase II trial of nivolumab plus ipilimumab. METHODS Key inclusion criteria included metastatic or recurrent adenocarcinoma of GC, ECOG performance status of 0 or 1, and no prior systemic therapy for metastatic or recurrent disease. MSI status was tested using multiplex PCR fragment analysis (MSI Testing Kit, FALCO). The primary endpoint was the prevalence of MSI-high GC. RESULTS Between October 2020 and October 2022, 930 eligible patients from 75 centers in Japan were analyzed. The prevalence of MSI-high GC was 5.6% (95% CI 4.2-7.3). MSI-high GC was more frequently observed in females than males (9.6% vs 3.8%, p < 0.001), patients aged ≥ 70 years compared to those < 70 years (8.0% vs 2.8%, p < 0.001), in the lower stomach than other locations (10.5% vs 3.2%, p < 0.001), HER2-negative tumors than HER2-positive tumors (6.5% vs 1.8%, p = 0.02), and in patients without liver metastasis than those with liver metastasis (6.9% vs 2.2%, p = 0.004). CONCLUSIONS The prevalence of MSI-high tumors among chemotherapy-naïve patients with unresectable GC was 5.6%. These tumors were associated with female sex, older age, lower stomach, HER2-negative, and absence of liver metastasis. These findings would help assuming MSI-high tumors and may have significant implications for clinical practice and studies targeting this GC subtype.
Collapse
Affiliation(s)
- Azusa Komori
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Shuichi Hironaka
- Department of Medical Oncology, Faculty of Medicine, Kyorin University, Mitaka, Japan.
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Seiichiro Mitani
- Department of Medical Oncology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Mitsuhiro Furuta
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Takeshi Kawakami
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | | | - Naoki Takegawa
- Department of Gastroenterology, Hyogo Cancer Center, Akashi, Japan
| | - Keiji Sugiyama
- Department of Medical Oncology, National Hospital Organization, Nagoya Medical Center, Nagoya, Japan
| | - Hidekazu Hirano
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tsukiji Campus, Chuo-Ku, Japan
| | - Takayuki Ando
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Tomohiro Matsushima
- Department of Gastroenterology, Saitama Prefectural Cancer Center, Ina, Japan
| | - Akihiko Chida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tomomi Kashiwada
- Department of Medical Oncology, Saga-Ken Medical Centre Koseikan, Saga, Japan
| | - Masato Komoda
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Toshihiko Matsumoto
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hisanobu Oda
- Division of Integrative Medical Oncology, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Hiroshi Yabusaki
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Hisato Kawakami
- Department of Medical Oncology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Narikazu Boku
- Department of Oncology and General Medicine, IMSUT Hospital, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ichinosuke Hyodo
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Kenichi Yoshimura
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| |
Collapse
|
16
|
de la Fouchardière C, Cammarota A, Svrcek M, Alsina M, Fleitas-Kanonnikoff T, Lordick Obermannová R, Wagner AD, Yap Wei Ting D, Enea D, Petrillo A, Smyth EC. How do I treat dMMR/MSI gastro-oesophageal adenocarcinoma in 2025? A position paper from the EORTC-GITCG gastro-esophageal task force. Cancer Treat Rev 2025; 134:102890. [PMID: 39933210 DOI: 10.1016/j.ctrv.2025.102890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
In less than a decade, immune checkpoint inhibitors (ICIs) have transformed the management of mismatch repair-deficient (dMMR) and microsatellite instability-high (MSI) cancers. However, beyond colorectal cancer (CRC), much of the evidence is mostly derived from non-randomized phase II studies or post-hoc analyses of broader clinical trials. dMMR/MSI tumours represent a specific subgroup of gastro-esophageal adenocarcinomas (GEA), accounting for approximately 9 % of cases, with a higher prevalence in early-stage compared to advanced-stage disease and older female patients. These tumours are predominantly sporadic, often linked to MLH1 promoter methylation, and rarely exhibit HER2 overexpression/ERBB2 amplification or other oncogenic drivers. The treatment landscape for early stage dMMR/MSI GEA is likely to change substantially soon, as ICIs have shown high pathological complete response (pCR) rates in small phase II trials, raising questions on optimisation of neoadjuvant therapy, and paving the way for organ preservation. The standard of treatment for untreated patients with advanced dMMR/MSI GEA is chemotherapy + ICI irrespectively of PDL-1 status. However, the role of chemotherapy-free regimen consisting of CTLA-4 plus PD-1 inhibitors remains undetermined. This review addresses these and other emerging questions, offering expert opinions and insights into the future therapeutic landscape for dMMR/MSI GEA.
Collapse
Affiliation(s)
- Christelle de la Fouchardière
- Institut PAOLI-CALMETTES, 232 Boulevard Sainte-Marguerite 13009, Marseille, France; Unicancer GI (UCGI) Group, Paris, France; EORTC-GITC Group, Brussels, Belgium.
| | - Antonella Cammarota
- EORTC-GITC Group, Brussels, Belgium; Hepatobiliary Immunopathology Lab, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Magali Svrcek
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Department of Pathology, France; LIMICS, UMRS 1142, Campus des Cordeliers 75006, Paris, France
| | - Maria Alsina
- EORTC-GITC Group, Brussels, Belgium; Hospital Universitario de Navarra, Navarrabiomed - IdiSNA, c. de Irunlarrea 3 31008, Pamplona, Spain
| | - Tania Fleitas-Kanonnikoff
- EORTC-GITC Group, Brussels, Belgium; Hospital Clínico Universitario de Valencia, INCLIVA, Valencia, Spain
| | - Radka Lordick Obermannová
- EORTC-GITC Group, Brussels, Belgium; Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of Medicine, Masaryk University, Czech Republic
| | - Anna Dorothea Wagner
- EORTC-GITC Group, Brussels, Belgium; Anna Dorothea Wagner, Department of Oncology, Division of Medical Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011, Lausanne, Switzerland
| | | | - Diana Enea
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Department of Pathology, France
| | - Angelica Petrillo
- EORTC-GITC Group, Brussels, Belgium; Medical Oncology Unit, Ospedale del Mare, Naples, Italy
| | - Elizabeth C Smyth
- EORTC-GITC Group, Brussels, Belgium; Oxford NIHRBiomedical Research Centre, Churchill Hospital, Oxford OX3 7LE, UK
| |
Collapse
|
17
|
Martínez-Ciarpaglini C, Barros R, Caballero C, Boggino H, Alarcón-Molero L, Peleteiro B, Ruiz-García E, Fernandez-Figueroa E, Herrera-Goepfert R, Díaz-Romero C, Ferreira R, Groen-van Schooten TS, Gauna C, Pereira R, Cantero D, Lezcano H, Esteso F, O Connor J, Riquelme A, Owen GI, Garrido M, Roa JC, Ruiz-Pace F, Vivancos A, Diez-García M, Alsina M, Matito J, Martin A, Gómez M, Castillo E, Vila M, Santos-Antunes J, Costa A, Lordick F, Farrés J, Palomar-De Lucas B, Cabeza-Segura M, Villagrasa R, Jimenez-Martí E, Miralles-Marco A, Dienstmann R, Derks S, Figueiredo C, Cervantes A, Carneiro F, Fleitas-Kanonnikoff T. Comprehensive histopathological analysis of gastric cancer in European and Latin America populations reveals differences in PDL1, HER2, p53 and MUC6 expression. Gastric Cancer 2025; 28:160-173. [PMID: 39755998 PMCID: PMC11842524 DOI: 10.1007/s10120-024-01578-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
INTRODUCTION Gastric cancer (GC) burden is currently evolving with regional differences associated with complex behavioural, environmental, and genetic risk factors. The LEGACy study is a Horizon 2020-funded multi-institutional research project conducted prospectively to provide comprehensive data on the tumour biological characteristics of gastroesophageal cancer from European and LATAM countries. MATERIAL AND METHODS Treatment-naïve advanced gastroesophageal adenocarcinoma patients were prospectively recruited in seven European and LATAM countries. Formalin-fixed paraffin-embedded primary tumour endoscopic biopsy samples were collected and submitted for central morphological and immunohistochemical characterization and TP53 molecular assessment and Helicobacter pylori infection. RESULTS A total of 259 patients were included in the study: 137 (53%) from LATAM and 122 (47%) from Europe. Significant biological differences were detected between European and LATAM patients. Low representation of chromosomal instability (CIN) and HER2 positive cases were found in LATAM. MUC6 and PD-L1 were more frequently overexpressed in European cases, showing a significant correlation across the entire study population, with this association being especially pronounced in MMRdeficient cases. Both TP53 mutation by next-generation sequencing and p53 immunohistochemical aberrant pattern were linked with features associated with chromosomal instability. No regional differences were observed in H. pylori prevalence or abundance, indicating that the afore mentioned variations cannot be attributed to this factor. CONCLUSION Our findings underscore a need for region-specific approaches in gastroesophageal cancer diagnosis and treatment. MUC6 emerges as a putative immune regulator that needs further investigation. Research tailored to the unique biological profiles in different global regions is crucial to effectively address the observed disparities.
Collapse
Affiliation(s)
- Carolina Martínez-Ciarpaglini
- Department of Pathology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Rita Barros
- Ipatimup, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Porto, Portugal
- Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Pathology, Unidade Local de Saúde São João, Porto, Portugal
| | | | - Hugo Boggino
- Department of Pathology, GENPAT, Asunción, Paraguay
| | - Lorena Alarcón-Molero
- Department of Pathology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia, Spain
- Department of Pathology, Hospital General de Valdepeñas, Valdepeñas, Spain
| | - Bárbara Peleteiro
- Hospital Epidemiology Center, University Hospital Center of São João, Porto, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
- EPIUnit-Institute of Public Health, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - Erika Ruiz-García
- Departamento de Tumores de Tubo Digestivo, Instituto Nacional de Cancerología, Mexico City, México
- Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerología, Mexico City, México
| | - Edith Fernandez-Figueroa
- Núcleo B de Innovación en Medicina de Precisión, Instituto Nacional de Medicina Genómica, Mexico City, México
| | | | - Consuelo Díaz-Romero
- Departamento de Oncología Médica, Instituto Nacional de Cancerología, Mexico City, México
| | - Rui Ferreira
- Ipatimup, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal
- Microbes & Cancer. i3S, Instituto de Investigação e Inovação em Saúde, , Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Tessa S Groen-van Schooten
- Department of Medical Oncology, Amsterdam University Medical Center (UMC) Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Cinthia Gauna
- Medical Oncology Department, Instituto de Previsión Social, Asunción, Paraguay
| | - Rita Pereira
- Medical Oncology Department, Instituto de Previsión Social, Asunción, Paraguay
| | - Daniel Cantero
- Department of Gastroenterology, Instituto de Previsión Social, Asunción, Paraguay
| | - Horacio Lezcano
- Pathology Department, Instituto de Previsión Social, Asunción, Paraguay
| | - Federico Esteso
- Medical Oncology Department, Instituto Alexander Fleming, Buenos Aires, Argentina
| | - Juan O Connor
- Medical Oncology Department, Instituto Alexander Fleming, Buenos Aires, Argentina
| | - Arnoldo Riquelme
- Department of Gastroenterology, Faculty of MedicineCenter for Prevention and Control of Cancer (CECAN), Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Gareth I Owen
- Faculty of Biological Sciences & Faculty of Medicine, Millennium Institute for Immunology and ImmunotherapyCenter for Prevention and Control of Cancer (CECAN), Advance Center for Chronic Disease (ACCDIS), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo Garrido
- Centro de Oncología de Precisión, Universidad Mayor, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology. Faculty of Medicine. Pontificia, Universidad Católica de Chile Santiago, Santiago, Chile
| | - Fiorella Ruiz-Pace
- Oncology Data Science, Valld`Hebron Institute of Oncology, Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Lab, Valld`Hebron Institute of Oncology, Barcelona, Spain
| | - Marc Diez-García
- Medical Oncology Department, Valld`Hebron Institute of Oncology, Barcelona, Spain
| | - Maria Alsina
- Medical Oncology Department, Valld`Hebron Institute of Oncology, Barcelona, Spain
- Hospital Universitario de Navarra, Navarrabiomed-IdiSNA, Pamplona, Spain
| | - Judit Matito
- Cancer Genomics Lab, Valld`Hebron Institute of Oncology, Barcelona, Spain
| | - Agatha Martin
- Cancer Genomics Lab, Valld`Hebron Institute of Oncology, Barcelona, Spain
| | - Marina Gómez
- Cancer Genomics Lab, Valld`Hebron Institute of Oncology, Barcelona, Spain
| | - Ester Castillo
- Cancer Genomics Lab, Valld`Hebron Institute of Oncology, Barcelona, Spain
| | - Maria Vila
- Cancer Genomics Lab, Valld`Hebron Institute of Oncology, Barcelona, Spain
| | - João Santos-Antunes
- Department of Gastroenterology, Unidade Local de Saúde São João, Porto, Portugal
| | - Andreia Costa
- Department of Oncology, Unidade Local de Saúde São João, Porto, Portugal
| | - Florian Lordick
- Department of Medicine (Oncology, Gastroenterology, Hepatology, and Pulmonology), Comprehensive Cancer Center Central Germany (CCCG), University of Leipzig Medical Center, Leipzig, Germany
| | | | - Brenda Palomar-De Lucas
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Avenida Menendez Pelayo nro 4 accesorio, Valencia, Spain
| | - Manuel Cabeza-Segura
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Avenida Menendez Pelayo nro 4 accesorio, Valencia, Spain
| | - Rosanna Villagrasa
- Department of Gastroenterology, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Elena Jimenez-Martí
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Avenida Menendez Pelayo nro 4 accesorio, Valencia, Spain
| | - Ana Miralles-Marco
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Avenida Menendez Pelayo nro 4 accesorio, Valencia, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science, Valld`Hebron Institute of Oncology, Barcelona, Spain
| | - Sarah Derks
- Department of Medical Oncology, Amsterdam University Medical Center (UMC) Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Ceu Figueiredo
- Ipatimup, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Porto, Portugal
- Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Andrés Cervantes
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Avenida Menendez Pelayo nro 4 accesorio, Valencia, Spain
- Department of Gastroenterology, Hospital Clínico Universitario de Valencia, Valencia, Spain
- CiberOnc. Carlos III Institute, Madrid, Spain
| | - Fátima Carneiro
- Ipatimup, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Porto, Portugal
- Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Pathology, Unidade Local de Saúde São João, Porto, Portugal
| | - Tania Fleitas-Kanonnikoff
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Avenida Menendez Pelayo nro 4 accesorio, Valencia, Spain.
| |
Collapse
|
18
|
Chen W, Wang H, Achi N, Hao J, Gong R, Zhao Q. Bioinformatics Analysis of the Expression and Prognostic Significance of Transcription Factor YY1 in Gastric Cancer. Cancer Rep (Hoboken) 2025; 8:e70181. [PMID: 40088083 PMCID: PMC11909590 DOI: 10.1002/cnr2.70181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Emerging evidence indicates that the transcription factor Yin Yang 1 (YY1) plays a critical role in the carcinogenesis and progression of various human malignancies. YY1 is highly expressed in gastric cancer (GC), raising interest in its role in GC. AIMS This study aims to analyze the role of YY1 in gastric cancer, investigate its effect on the tumor microenvironment, and assess its potential as a prognostic marker. METHODS AND RESULTS Transcriptomic data and clinical information from GC patients were obtained from the TCGA and UCSC databases. YY1 expression was analyzed using the R "limma" package. Gene ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed with the online tool clusterProfiler. The relationship between YY1 expression levels and the tumor microenvironment was examined in different risk groups of GC patients. Additionally, YY1-positive staining in 26 clinical GC samples was measured using ImageJ software. Co-expression analysis was used to identify prognostic genes associated with YY1, and a prognostic risk model was built and optimized. Results showed that YY1 was significantly overexpressed in 415 GC (p < 0.001) and was associated with poorer survival outcomes (p = 0.043). GO and KEGG showed that YY1 was involved in key biological processes of the disease. Higher YY1 expression was correlated with lower stromal and immune cell content in the tumor microenvironment. Immunohistochemical staining confirmed YY1 overexpression in GC tissues compared to normal tissues (p = 0.0293). Positive correlations were observed between YY1 and the genes MTA1, TTL15, HNRNPU, WDR20, and PPP4R3A. The prognostic model, which included genes significantly associated with YY1 (risk score AUC = 0.690), predicted patient survival better than other clinical variables. CONCLUSION These findings suggest that YY1 plays an important role in the development of GC. Targeting the YY1 pathway may be a potential treatment strategy for GC.
Collapse
Affiliation(s)
- Wenliang Chen
- Department of General Surgery, The 2nd Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huanhuan Wang
- Department of General Surgery, Jincheng People's Hospital, Jincheng, Shanxi, China
| | - Ntiak Achi
- Graduate Department of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinjin Hao
- Graduate Department of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Gong
- Graduate Department of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qiang Zhao
- Graduate Department of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
19
|
Mahmoudian-Hamedani S, Lotfi-Shahreza M, Nikpour P. Investigating combined hypoxia and stemness indices for prognostic transcripts in gastric cancer: Machine learning and network analysis approaches. Biochem Biophys Rep 2025; 41:101897. [PMID: 39807391 PMCID: PMC11729012 DOI: 10.1016/j.bbrep.2024.101897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction Gastric cancer (GC) is among the deadliest malignancies globally, characterized by hypoxia-driven pathways that promote cancer progression, including stemness mechanisms facilitating invasion and metastasis. This study aimed to develop a prognostic decision tree using genes implicated in hypoxia and stemness pathways to predict outcomes in GC patients. Materials and methods GC RNA-seq data from The Cancer Genome Atlas (TCGA) were analyzed to compute hypoxia and stemness scores using Gene Set Variation Analysis (GSVA) and the mRNA expression-based stemness index (mRNAsi). Hierarchical clustering identified clusters with distinct survival outcomes, and differentially expressed genes (DEGs) between clusters were identified. Weighted Gene Co-expression Network Analysis (WGCNA) identified modules and hub genes associated with clinical traits. Overlapping DEGs and hub genes underwent functional enrichment, protein-protein interaction (PPI) network analysis, and survival analysis. A prognostic decision tree was constructed using survival-associated shared genes. Results Hierarchical clustering identified six clusters among 375 TCGA GC patients, with significant survival differences between cluster 1 (low hypoxia, high stemness) and cluster 4 (high hypoxia, high stemness). Validation in the GSE62254 dataset corroborated these findings. WGCNA revealed modules linked to clinical traits and survival, with functional enrichment highlighting pathways like cell adhesion and calcium signaling. The decision tree, based on genes such as AKAP6, GLRB, and RUNX1T1, achieved an AUC of 0.81 (training) and 0.67 (test), demonstrating the utility of combined scores in patient stratification. Conclusion This study introduces a novel hypoxia-stemness-based prognostic decision tree for GC. The identified genes show promise as prognostic biomarkers, warranting further clinical validation.
Collapse
Affiliation(s)
- Sharareh Mahmoudian-Hamedani
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Lotfi-Shahreza
- Department of Computer Engineering, Shahreza Campus, University of Isfahan, Isfahan, Iran
| | - Parvaneh Nikpour
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
20
|
Dienstmann R, Ruiz-García E, Alsina M, Ruiz-Pace F, Groen-van Schooten TS, Martínez-Ciarpaglini C, Fernández-Figueroa EA, Herrera-Goepfert R, Díaz-Romero C, Lino-Silva L, Hernandez-Guerrero AI, Valdez-Reyes NM, León-Takahashi A, Falcón-Martínez JC, Pouw RE, Romero S, Villagrasa R, Cabeza-Segura M, Alarcón-Molero L, Jimenez-Martí E, Miralles A, Boggino H, Gauna C, Pereira R, Lezcano H, Cantero D, Vivancos A, Matito J, Martin A, Gómez M, Castillo E, Vila M, Ferreira RM, Barros R, Santos-Antunes J, Mendes-Rocha M, Costa A, Riquelme E, Roa JC, Latorre G, Freile B, Caro L, Esteso F, O'Connor J, Riquelme A, Owen G, Garrido M, Diez-García M, Figueiredo C, Caballero C, Lordick F, Farrés J, Derks S, Carneiro F, Cervantes A, Fleitas T. Integrated clinico-molecular analysis of gastric cancer in European and Latin American populations: LEGACY project. ESMO Open 2025; 10:104482. [PMID: 40036904 PMCID: PMC11926697 DOI: 10.1016/j.esmoop.2025.104482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is recognized for intrinsic heterogeneity, although it is similarly approached in Europe and Latin America (LATAM). The LEGACY project aimed to deepen GC molecular understanding through multi-omics analysis in Europe and LATAM GC samples. PATIENTS AND METHODS Tumor samples were centrally reviewed for histology, human epidermal growth factor receptor 2 (HER2) expression, and mismatch repair-deficient (dMMR)/microsatellite instability (MSI) status. In addition, we assessed Epstein-Barr virus (EBV) status, programmed death-ligand 1 (PD-L1) combined positive score (CPS), and carried out tissue genomic profiling including tumor mutation burden (TMB) quantification plus targeted transcriptomics for immune microenvironment and cancer cell signaling scores. RESULTS In total, 328 GC patients were enrolled. HER2-positive GC and high PD-L1 CPS were more frequent in Europe than in LATAM (9% versus 3% and 15% versus 3%, respectively), whereas EBV was mainly found in LATAM (7%, versus 3% in Europe), and dMMR/MSI tumors were equally distributed (16%). High TMB was enriched in dMMR/MSI and EBV tumors. Mutations in homologous recombination repair (HRR) genes were frequent in both cohorts (24.8% and 14.7% in Europe and LATAM, respectively), and mostly found in dMMR/MSI (63.6%) and intestinal HER2-negative (18.7%) tumors. The prognosis was poor in diffuse HER2-negative GC patients, whose tumors presented an immunosuppressive microenvironment and other distinct pathway activation signatures. CONCLUSIONS Our findings relate specific molecular alterations of GC tumors from Europe and LATAM to actionable biomarkers for precision cancer therapies. The proposed GC stratification can be implemented in routine care and guide drug development strategies.
Collapse
Affiliation(s)
- R Dienstmann
- Oncology Data Science, Vall d´Hebron Institute of Oncology, Barcelona, Spain; OC Precision Medicine, Oncoclínicas & Co, São Paulo, Brazil; University of Vic-Central University of Catalonia, Barcelona, Spain. https://twitter.com/rdienstmann
| | - E Ruiz-García
- Departamento de Tumores de Tubo Digestivo, Instituto Nacional de Cancerología, Mexico City, Mexico; Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerología, Mexico City, Mexico. https://twitter.com/Ruiz-García
| | - M Alsina
- Medical Oncology Department, Vall d`Hebron Institute of Oncology, Barcelona, Spain; Hospital Universitario de Navarra, Navarrabiomed-IdiSNA, Pamplona, Spain. https://twitter.com/Alsina
| | - F Ruiz-Pace
- Oncology Data Science, Vall d´Hebron Institute of Oncology, Barcelona, Spain. https://twitter.com/Ruiz-Pace
| | - T S Groen-van Schooten
- Department of Medical Oncology, Amsterdam University Medical Center (UMC), location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - C Martínez-Ciarpaglini
- Department of Pathology, Hospital Clínico Universitario de Valencia, Valencia, Spain. https://twitter.com/Martínez-Ciarpaglini
| | - E A Fernández-Figueroa
- Núcleo B de Innovación en Medicina de Precisión, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - R Herrera-Goepfert
- Department of Pathology, Instituto Nacional de Cancerología, Mexico City, Mexico. https://twitter.com/Herrera-Goepfert
| | - C Díaz-Romero
- Department of Medical Oncology, Instituto Nacional de Cancerología, Mexico City, Mexico. https://twitter.com/Díaz-Romero
| | - L Lino-Silva
- Department of Head of Division, Surgical Pathology, National Cancer Institute (INCan), Mexico City, Mexico. https://twitter.com/Lino-Silva
| | - A I Hernandez-Guerrero
- Department of Gastrointestinal Endoscopy, Instituto Nacional de Cancerología, Mexico City, Mexico. https://twitter.com/Hernandez-Guerrero
| | - N M Valdez-Reyes
- Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - A León-Takahashi
- Departamento de Gastroenterología, Instituto Nacional de Cancerología, Mexico City, Mexico. https://twitter.com/León-Takahashi
| | - J C Falcón-Martínez
- Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - R E Pouw
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center (UMC), location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - S Romero
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia, Spain. https://twitter.com/Romero
| | - R Villagrasa
- Department of Gastroenterology, Hospital Clínico Universitario de Valencia, Valencia, Spain. https://twitter.com/Villagrasa
| | - M Cabeza-Segura
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia, Spain. https://twitter.com/Cabeza-Segura
| | - L Alarcón-Molero
- Department of Pathology, Hospital Clínico Universitario de Valencia, Valencia, Spain; Department of Pathology, Hospital General de Valdepeñas, Valdepeñas, Spain. https://twitter.com/Alarcón-Molero
| | - E Jimenez-Martí
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia, Spain. https://twitter.com/Jimenez-Martí
| | - A Miralles
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - H Boggino
- Department of Pathology, GENPAT, Asunción, Paraguay
| | - C Gauna
- Department of Medical Oncology, Instituto de Previsión Social, Asunción, Paraguay
| | - R Pereira
- Department of Medical Oncology, Instituto de Previsión Social, Asunción, Paraguay
| | - H Lezcano
- Department of Pathology, Instituto de Previsión Social, Asunción, Paraguay
| | - D Cantero
- Department of Gastroenterology, Instituto de Previsión Social, Asunción, Paraguay
| | - A Vivancos
- Cancer Genomics Lab, Vall d`Hebron Institute of Oncology, Barcelona, Spain. https://twitter.com/Vivancos
| | - J Matito
- Cancer Genomics Lab, Vall d`Hebron Institute of Oncology, Barcelona, Spain. https://twitter.com/Matito
| | - A Martin
- Cancer Genomics Lab, Vall d`Hebron Institute of Oncology, Barcelona, Spain. https://twitter.com/Martin
| | - M Gómez
- Cancer Genomics Lab, Vall d`Hebron Institute of Oncology, Barcelona, Spain. https://twitter.com/Gómez
| | - E Castillo
- Cancer Genomics Lab, Vall d`Hebron Institute of Oncology, Barcelona, Spain. https://twitter.com/Castillo
| | - M Vila
- Cancer Genomics Lab, Vall d`Hebron Institute of Oncology, Barcelona, Spain. https://twitter.com/Vila
| | - R M Ferreira
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. https://twitter.com/Ferreira
| | - R Barros
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Faculty of Medicine of the University of Porto, Porto, Portugal; Department of Pathology, Unidade Local de Saúde São João, Porto, Portugal
| | - J Santos-Antunes
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Faculty of Medicine of the University of Porto, Porto, Portugal; Department of Gastroenterology, Unidade Local de Saúde São João, Porto, Portugal. https://twitter.com/Santos-Antunes
| | - M Mendes-Rocha
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Faculty of Medicine of the University of Porto, Porto, Portugal. https://twitter.com/Mendes-Rocha
| | - A Costa
- Department of Oncology, Unidade Local de Saúde São João, Porto, Portugal
| | - E Riquelme
- Department of Respiratory Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J C Roa
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - G Latorre
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - B Freile
- Department of Medical Oncology Department, Instituto Alexander Fleming, Buenos Aires, Argentina
| | - L Caro
- Department of Gastroenterology, Instituto Alexander Fleming, GEDYT (Gastroenterologia diagnostica y terapeutica), Buenos Aires, Argentina
| | - F Esteso
- Department of Medical Oncology Department, Instituto Alexander Fleming, Buenos Aires, Argentina. https://twitter.com/federico_esteso
| | - J O'Connor
- Department of Medical Oncology Department, Instituto Alexander Fleming, Buenos Aires, Argentina. https://twitter.com/juanmaoconnor
| | - A Riquelme
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Center for Prevention and Control of Cancer (CECAN), Santiago, Chile
| | - G Owen
- Faculty of Biological Sciences & Faculty of Medicine, Pontificia Universidad Católica de Chile, Millennium Institute for Immunology and Immunotherapy, Center for Prevention and Control of Cancer (CECAN), Advance Center for Chronic Disease (ACCDIS), Santiago, Chile
| | - M Garrido
- Facultad de Medicina y Ciencia de la Salud, Centro de Oncología de Precision, Universidad Mayor, Santiago, Chile. https://twitter.com/DrGarridoOncoGI
| | - M Diez-García
- Medical Oncology Department, Vall d`Hebron Institute of Oncology, Barcelona, Spain. https://twitter.com/Diez-García
| | - C Figueiredo
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Faculty of Medicine of the University of Porto, Porto, Portugal. https://twitter.com/FigeuiredoCeu
| | - C Caballero
- Department of Pathology, GENPAT, Asunción, Paraguay
| | - F Lordick
- Department of Oncology and University Cancer Center Leipzig, University of Leipzig Medical Center, Leipzig, Germany. https://twitter.com/FlorianLordick
| | - J Farrés
- Anaxomics Biotech S.L., Barcelona, Spain
| | - S Derks
- Department of Medical Oncology, Amsterdam University Medical Center (UMC), location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands. https://twitter.com/derks_s
| | - F Carneiro
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Faculty of Medicine of the University of Porto, Porto, Portugal; Department of Pathology, Unidade Local de Saúde São João, Porto, Portugal. https://twitter.com/Carneiro
| | - A Cervantes
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia, Spain; Ciberonc, Instituto Carlos III, Madrid, Spain.
| | - T Fleitas
- Department of Medical Oncology, Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia, Spain; Ciberonc, Instituto Carlos III, Madrid, Spain.
| |
Collapse
|
21
|
Chen C, Shao Y, Ye C, Yu X, Hu M, Yan J, Ye G. Weighted Gene Coexpression Network Analysis Identifies Neutrophil-Related Molecular Subtypes and Their Clinical Significance in Gastric Cancer. Cancer Manag Res 2025; 17:397-418. [PMID: 40040634 PMCID: PMC11878151 DOI: 10.2147/cmar.s500215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/22/2025] [Indexed: 03/06/2025] Open
Abstract
Background Gastric cancer (GC) is among the most lethal malignancies worldwide. Due to the substantial heterogeneity of GC, more accurate molecular typing systems are desperately required to enhance the prognosis of GC patients. Methods The major immune cell subclusters in GC were identified by a single-cell RNA sequencing (scRNA-seq) dataset. High-dimensional weighted gene coexpression network analysis (hdWGCNA) and multiple bioinformatics methods were utilized to classify the molecular subtypes of GC and further investigate the differences among the subtypes. Based on the module genes and differentially expressed genes (DEGs), random survival forest analysis was applied to identify the key prognostic genes for GC, and the roles and functional mechanisms of the key genes in GC were explored by clinical samples and cellular experiments. Results Two distinct GC molecular subtypes (C1 and C2) associated with neutrophils were identified, with C1 associated with better prognosis. Compared with C2 subtype, C1 subtype has significant differences in immune infiltration, immune checkpoint expression, signaling pathway regulation, tumor mutation burden, and immunotherapy and chemotherapeutic drug sensitivity. Three new key genes (VIM, RBMS1 and RGS2) were revealed to be highly correlated with the prognosis of GC patients. In addition, the expression and cellular functions of key genes RBMS1 and RGS2 in gastric carcinogenesis were verified. Conclusion We identified two neutrophil-related molecular GC subtypes with different prognostic outcomes and clinical significance. VIM, RBMS1 and RGS2 were identified as potential prognostic markers and therapeutic targets for GC. These findings provide a new perspective for the molecular typing and personalized treatment of GC.
Collapse
Affiliation(s)
- Chujia Chen
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, People’s Republic of China
- Health Science Center, Ningbo University, Ningbo, 315211, People’s Republic of China
| | - Yongfu Shao
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, People’s Republic of China
| | - Chengyuan Ye
- Health Science Center, Ningbo University, Ningbo, 315211, People’s Republic of China
| | - Xuan Yu
- Health Science Center, Ningbo University, Ningbo, 315211, People’s Republic of China
| | - Meng Hu
- Health Science Center, Ningbo University, Ningbo, 315211, People’s Republic of China
| | - Jianing Yan
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, People’s Republic of China
| | - Guoliang Ye
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, People’s Republic of China
| |
Collapse
|
22
|
Lewis KA, Diggs LP, Badgwell BD. Educational Review: Updates on Therapeutic Strategies for Gastric Cancer with Peritoneal Metastasis. Ann Surg Oncol 2025:10.1245/s10434-025-17069-3. [PMID: 40016614 DOI: 10.1245/s10434-025-17069-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 02/09/2025] [Indexed: 03/01/2025]
Abstract
Gastric cancer (GC) commonly presents in advanced stages with metastatic spread to the peritoneal cavity, and outcomes associated with gastric cancer with peritoneal metastasis (GCPM) continue to carry a dismal prognosis. Persistent challenges in the detection of peritoneal metastasis (PM) have resulted in a relative paucity of high-quality data to inform management decisions. Several consensus groups have published recommendations to guide management, including most recently the National Comprehensive Cancer Network guidelines, which now include cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) as a potential treatment modality in select patients with GCPM. Multiple clinical trials have investigated the use of CRS/HIPEC and other peritoneal-directed therapies, such as intraperitoneal chemotherapy (IPC) and pressurized intraperitoneal aerosolized chemotherapy (PIPAC). As high-volume centers work to incorporate such therapies into their practice, ongoing clinical trials are aimed at understanding their efficacy. Recent findings have improved understanding of the mechanisms and pathophysiology underlying GCPM while the discovery of novel targets offers potential for drug development and therapeutic strategies to overcome treatment resistance. This review highlights recent advancements and addresses the persistent challenges in managing GCPM while also offering a comprehensive summary of current guidelines and treatment strategies.
Collapse
Affiliation(s)
- Kever A Lewis
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Laurence P Diggs
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian D Badgwell
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Semenova Y, Kerimkulov A, Uskenbayev T, Zharlyganova D, Shatkovskaya O, Sarina T, Manatova A, Yessenbayeva G, Adylkhanov T. Chemotherapy Options for Locally Advanced Gastric Cancer: A Review. Cancers (Basel) 2025; 17:809. [PMID: 40075656 PMCID: PMC11899121 DOI: 10.3390/cancers17050809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Cancers represent a significant global health burden, affecting millions of individuals each year [...].
Collapse
Affiliation(s)
- Yuliya Semenova
- Department of Surgery, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan;
| | - Altay Kerimkulov
- Department of Multidisciplinary Surgery, National Research Oncology Center, Astana 020000, Kazakhstan; (A.K.); (T.U.); (T.S.); (T.A.)
| | - Talgat Uskenbayev
- Department of Multidisciplinary Surgery, National Research Oncology Center, Astana 020000, Kazakhstan; (A.K.); (T.U.); (T.S.); (T.A.)
| | - Dinara Zharlyganova
- Department of Scientific Management, National Research Oncology Center, Astana 020000, Kazakhstan; (D.Z.); (G.Y.)
| | - Oxana Shatkovskaya
- Board for Strategic Development, Scientific and Educational Activities, National Research Oncology Center, Astana 020000, Kazakhstan;
| | - Tomiris Sarina
- Department of Multidisciplinary Surgery, National Research Oncology Center, Astana 020000, Kazakhstan; (A.K.); (T.U.); (T.S.); (T.A.)
| | - Almira Manatova
- Department of Scientific Management, National Research Oncology Center, Astana 020000, Kazakhstan; (D.Z.); (G.Y.)
| | - Gulfairus Yessenbayeva
- Department of Scientific Management, National Research Oncology Center, Astana 020000, Kazakhstan; (D.Z.); (G.Y.)
| | - Tasbolat Adylkhanov
- Department of Multidisciplinary Surgery, National Research Oncology Center, Astana 020000, Kazakhstan; (A.K.); (T.U.); (T.S.); (T.A.)
| |
Collapse
|
24
|
Zhang H, Hu J, Li Y, Liu Y, Shen H, Wang Z, Li Q. Comprehensive analysis and experimental validation of disulfidptosis-associated prognostic signature and immune microenvironment characterization of gastric cancer. Cancer Immunol Immunother 2025; 74:116. [PMID: 39998563 PMCID: PMC11861452 DOI: 10.1007/s00262-024-03883-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/03/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common causes of cancer-related death worldwide. As a novel form of programmed cell death, disulfidptosis is characterized by excessive cysteine accumulation, disulfide stress and actin destruction. There is evidence that targeting disulfidptosis is a promising anticancer strategy. Further improvement of GC risk stratification based on disulfidptosis has positive clinical significance. METHODS We analyzed the expression levels of disulfidptosis-associated genes (DPAGs) in normal and GC tissues and characterized the molecular subtypes of GC patients. Based on the characteristics of DPAG subtypes, differentially expressed prognosis-related genes were selected by LASSO-univariate Cox analysis and multivariate Cox analysis analyzed to establish a prognostic model. Using single-cell sequencing analysis reveals the cell subpopulation for GC. The function of the selected target in GC was verified by in vitro experimental means, including siRNA, qRT-PCR, Western blot, CCK-8, and Transwell assay. RESULTS DPAG score was verified to be an independent prognostic factor of GC and was significantly associated with poor prognosis of gastric cancer. Subsequent studies on subgroup immunoinfiltration characteristics, drug sensitivity analysis, immunotherapy response and somatic mutation characteristics of DPAG score comprehensively confirmed the potential guiding significance of DPAG score for individualized treatment of gastric cancer patients. Single-cell sequencing analysis revealed the expression characteristics of DPAG-related prognostic signatures across cell subpopulations. In vitro experiments showed APC11, as one of the selected DPAGs, was highly expressed in gastric cancer, and knockdown of APC11 could significantly inhibit the proliferation and migration of GC cells, demonstrating the reliability of bioinformatics results. CONCLUSION The results of this study provide a new perspective for exploring the role of disulfidptosis in the occurrence and development of GC.
Collapse
Affiliation(s)
| | - Jinguo Hu
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yuanqiang Li
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yanyang Liu
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Huize Shen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zeng Wang
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Qinglin Li
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China.
| |
Collapse
|
25
|
Jiang J, Chen Y, Zheng Y, Ding Y, Wang H, Zhou Q, Teng L, Zhang X. Sialic acid metabolism-based classification reveals novel metabolic subtypes with distinct characteristics of tumor microenvironment and clinical outcomes in gastric cancer. Cancer Cell Int 2025; 25:61. [PMID: 39987095 PMCID: PMC11847363 DOI: 10.1186/s12935-025-03695-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/13/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND High heterogeneity in gastric cancer (GC) remains a challenge for standard treatments and prognosis prediction. Dysregulation of sialic acid metabolism (SiaM) is recognized as a key metabolic hallmark of tumor immune evasion and metastasis. Herein, we aimed to develop a SiaM-based metabolic classification in GC. METHODS SiaM-related genes were obtained from the MsigDB database. Bulk and single-cell transcriptional data of 956 GC patients were acquired from the GEO, TCGA, and MEDLINE databases. Proteomic profiles of 20 GC samples were derived from our institution. The consensus clustering algorithm was applied to identify SiaM-based clusters. The SiaM-based model was established via LASSO regression and evaluated via Kaplan‒Meier curve and ROC curve analyses. In vitro and in vivo experiments were conducted to explore the function of ST3GAL1 in GC. RESULTS Three SiaM clusters presented distinct patterns of clinicopathological features, transcriptomic alterations, and tumor immune microenvironment landscapes in GC. Compared with clusters A and B, cluster C presented elevated SiaM activity, higher metastatic potential, more abundant immunosuppressive features, and a worse prognosis. Based on the differentially expressed genes between these clusters, a risk model for six genes (ARHGAP6, ST3GAL1, ADAM28, C7, PLCL1, and TTC28) was then constructed. The model exhibited robust performance in predicting peritoneal metastasis and prognosis in four independent cohorts. As a hub gene in the model, ST3GAL1 promoted GC cell migration and invasion in vitro and in vivo. CONCLUSIONS Our study proposed a novel SiaM-based classification that identified three metabolic subtypes with distinct characteristics of tumor microenvironment and clinical outcomes in GC.
Collapse
Affiliation(s)
- Junjie Jiang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Gastroenterology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Hangzhou Institute of Digestive Disease, Hangzhou, Zhejiang, China
| | - Yiran Chen
- Department of Surgical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yangyang Zheng
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yongfeng Ding
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haiyong Wang
- Department of Surgical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Quan Zhou
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Surgical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lisong Teng
- Department of Surgical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, 261 Huansha Road, Hangzhou, 310006, Zhejiang, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China.
- Hangzhou Institute of Digestive Disease, Hangzhou, Zhejiang, China.
| |
Collapse
|
26
|
Chen Y, Liu D, Wei K, Lin Y, Wang Z, Sun Q, Wang H, Peng J, Lian L. Carcinoembryonic antigen trajectory predicts pathological complete response in advanced gastric cancer after neoadjuvant chemotherapy. Front Oncol 2025; 15:1525324. [PMID: 39995833 PMCID: PMC11847669 DOI: 10.3389/fonc.2025.1525324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Aims This study aims to develop a simple, clinically applicable classification system to predict pCR based on carcinoembryonic antigen (CEA) trajectory during NAC. Methods This study included 366 AGC patients who received NAC followed by radical gastrectomy. CEA levels were measured before, during, and after NAC, with changes classified into three trajectory types: Type I (>=80% decline), Type II (>=40% but <80% decline), and Type III (<40% decline or increase). We analyzed associations between these CEA trajectories, pCR, lymph node remission, and survival. Results pCR was achieved in 10.4% (38/366) of patients. pCR rates were significantly higher in Type I (41%) and Type II (15.8%) trajectories compared to Type III (6.7%). Lymph node remission also correlated with CEA trajectories, with Type I having the highest proportion of ypN0 (79.2%). Multivariate analysis identified CEA trajectory subtypes and tumor differentiation as independent predictors of pCR. This classification system proved robust across subgroups. Although no significant differences in overall survival were observed between subtypes, higher initial CEA levels were associated with worse survival. Conclusion The trajectory of CEA change during NAC is a promising predictor of pCR in AGC. This simple and accessible classification system may facilitate personalized surgical strategies for patients with AGC.
Collapse
Affiliation(s)
- Yonghe Chen
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dan Liu
- Department of Laboratory Science, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaikai Wei
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Lin
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Wang
- School of Nursing, Sun Yat-sen University, Guangzhou, China
| | - Qian Sun
- School of Nursing, Sun Yat-sen University, Guangzhou, China
| | - Huashe Wang
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junsheng Peng
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Lian
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Zan L, Zhang X, Shen L, Zhao Q, Tan D, Peng X, Jia Y, Li J, Liu J, Zhao J, Gao N, Bu P, Xi Y. Genomic landscape and potential therapeutic targets in alpha-fetoprotein-producing gastric cancer. Gastric Cancer 2025:10.1007/s10120-025-01594-x. [PMID: 39928247 DOI: 10.1007/s10120-025-01594-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/11/2025]
Abstract
Alpha-fetoprotein-producing gastric carcinoma (AFPGC) is a rare and aggressive subtype of gastric cancer (GC). A comprehensive analysis of clinicopathological features, immunophenotypes, molecular characteristics, and survival in AFPGC contributes to identifying potential therapeutic targets and developing new strategies to manage this disease. A retrospective cohort study was conducted at Shanxi Cancer Hospital from January 2018 to December 2020, involving patients diagnosed with GC and elevated AFP serum levels. Among these, 91 patients underwent immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and next-generation sequencing (NGS) to reveal the immunophenotypic and molecular characteristics of AFPGC. We found that AFPGC is more common in males and primarily occurs in the cardia and antrum of the stomach. A panel of IHC markers including AFP, GPC3, SALL4, CD10, CDX-2, and ATBF1 can be used for the diagnosis and differentiating AFPGC. NGS analysis revealed that TP53 hypermutation was the most frequent molecular event associated with AFPGC. The altered signaling pathways included disease signal transduction, receptor tyrosine kinase signaling and intracellular second messenger signaling pathways. The cumulative incidence of 21 genes, based on the evidence of clinical actionability in the OncoKB, was found to be 59.34%. Among these genes, CCNE1, ERBB2, and EGFR were the most frequently observed. This underscores the potential benefit of targeted therapy for patients with AFPGC. Furthermore, LRP1B and ARID1A have been identified as prognostic factors associated with overall survival (OS) and disease-free survival (DFS), respectively. Our results aim to improve AFPGC diagnosis by identifying potential therapeutic targets and prognostic factors, which could help facilitate the development of new treatment strategies.
Collapse
Affiliation(s)
- Likun Zan
- Department of Pathology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xin Zhang
- Department of Colorectal Surgery, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lulu Shen
- Department of Pathology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Pathology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Qi Zhao
- Department of Pathology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Dongfeng Tan
- Departments of Pathology and Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiao Peng
- Department of Pathology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Pathology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Sichuan, China
| | - Yi Jia
- Department of Gastrointestinal Surgery, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiawen Li
- Department of Pathology, Basic Medicine, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jing Liu
- Department of Pathology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiaqi Zhao
- Department of Pathology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Pathology, Basic Medicine, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Ning Gao
- Department of Pathology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Peng Bu
- Department of Pathology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanfeng Xi
- Department of Pathology, Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
28
|
Shimura M, Matsuo J, Pang S, Jangphattananont N, Hussain A, Rahmat MB, Lee JW, Douchi D, Tong JJL, Myint K, Srivastava S, Teh M, Koh V, Yong WP, So JBY, Tan P, Yeoh KG, Unno M, Chuang LSH, Ito Y. IQGAP3 signalling mediates intratumoral functional heterogeneity to enhance malignant growth. Gut 2025; 74:364-386. [PMID: 39438124 PMCID: PMC11874294 DOI: 10.1136/gutjnl-2023-330390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/05/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND The elevation of IQGAP3 expression in diverse cancers indicates a key role for IQGAP3 in carcinogenesis. Although IQGAP3 was established as a proliferating stomach stem cell factor and a regulator of the RAS-ERK pathway, how it drives cancer growth remains unclear. OBJECTIVE We define the function of IQGAP3 in gastric cancer (GC) development and progression. DESIGN We studied the phenotypic changes caused by IQGAP3 knockdown in three molecularly diverse GC cell lines by RNA-sequencing. In vivo tumorigenesis and lung metastasis assays corroborated IQGAP3 as a mediator of oncogenic signalling. Spatial analysis was performed to evaluate the intratumoral transcriptional and functional differences between control tumours and IQGAP3 knockdown tumours. RESULTS Transcriptomic profiling showed that IQGAP3 inhibition attenuates signal transduction networks, such as KRAS signalling, via phosphorylation blockade. IQGAP3 knockdown was associated with significant inhibition of MEK/ERK signalling-associated growth factors, including TGFβ1, concomitant with gene signatures predictive of impaired tumour microenvironment formation and reduced metastatic potential. Xenografts involving IQGAP3 knockdown cells showed attenuated tumorigenesis and lung metastasis in immunodeficient mice. Accordingly, immunofluorescence staining revealed significant reductions of TGFβ/SMAD signalling and αSMA-positive stromal cells; digital spatial analysis indicated that IQGAP3 is indispensable for the formation of two phenotypically diverse cell subpopulations, which played crucial but distinct roles in promoting oncogenic functions. CONCLUSION IQGAP3 knockdown suppressed the RAS-TGFβ signalling crosstalk, leading to a significant reduction of the tumour microenvironment. In particular, IQGAP3 maintains functional heterogeneity of cancer cells to enhance malignant growth. IQGAP3 is thus a highly relevant therapy target in GC.
Collapse
Affiliation(s)
- Mitsuhiro Shimura
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junichi Matsuo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - ShuChin Pang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Aashiq Hussain
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Jung-Won Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Daisuke Douchi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jasmine Jie Lin Tong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Khine Myint
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Ming Teh
- Department of Medicine, National University of Singapore, Singapore
| | - Vivien Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- National University Cancer Institute, National University Health System, Singapore
| | - Wei Peng Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- National University Cancer Institute, National University Health System, Singapore
| | - Jimmy Bok Yan So
- Department of Surgery, National University of Singapore, Singapore
| | - Patrick Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Duke-NUS Graduate Medical School, Singapore
- Genome Institute of Singapore, Singapore
| | - Khay-Guan Yeoh
- Department of Medicine, National University of Singapore, Singapore
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Yoshiaki Ito
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| |
Collapse
|
29
|
Balmaceda NB, Kim SS. Evolving Strategies in the Management of Microsatellite Instability-High/Mismatch Repair Deficient Esophagogastric Adenocarcinoma. Curr Oncol Rep 2025; 27:81-94. [PMID: 39832053 DOI: 10.1007/s11912-024-01624-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE OF REVIEW This review addresses the current treatment paradigm and new advancements in the management of microsatellite instability-high/mismatch repair deficient (MSI-H/dMMR) esophagogastric cancer (EGC). RECENT FINDINGS While chemotherapy and surgery remain the cornerstone of EGC treatment, MSI-H/dMMR tumors harbor high tumor mutational burden and represent a subset of patients who benefit from immune checkpoint inhibitors (ICI). ICI has been incorporated in the front line setting with and without chemotherapy for advanced disease. Recently, ICI has been studied in the perioperative setting for resectable disease. Though perioperative ICI results in improved response rates, it is not yet clear whether this translates to a survival benefit. Despite high response rates with ICI in this patient population, many do not respond to therapy, representing a major challenge in treatment. Preclinical studies have highlighted potential mechanisms of resistance which will guide drug development and clinical trials.
Collapse
Affiliation(s)
- Nicole Baranda Balmaceda
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sunnie S Kim
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
30
|
Pettigrew MF, Al Abbas AI, Southichack A, Ju MR, Hammer STG, Liu Y, Porembka MR, Wang SC. Factors Associated With Minority Patient Enrollment in a Gastric Cancer Biobank. J Surg Res 2025; 306:230-238. [PMID: 39798410 PMCID: PMC11911076 DOI: 10.1016/j.jss.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/16/2024] [Accepted: 12/01/2024] [Indexed: 01/15/2025]
Abstract
INTRODUCTION Human tissue samples are essential for translational cancer research. However, less than 20% of current biobank and genomic samples were obtained from minority patients, which may lead to biased understanding of cancer biology. The objective of this study was to identify factors associated with patient enrollment in our institution's gastric cancer biobank. METHODS Patients with suspected or confirmed gastric or gastroesophageal junction cancer undergoing surgical procedures at our institution were invited to enroll in a prospective gastric cancer biobank. We retrospectively reviewed patients who were invited to enroll from 2017 to 2023 at our safety-net and university hospitals. We compared patients who enrolled to those who declined to identify factors that predict enrollment. RESULTS Hispanic patients had similar odds of enrollment as non-Hispanic White patients (odds ratio (OR): 1.22, 95% confidence interval (CI): 0.54-2.73, P = 0.63). Non-Hispanic minorities (Black/African Americans and Asians) were less likely to enroll when compared to non-Hispanic Whites (OR: 0.41, 95% CI: 0.18-0.95, P = 0.04). Minority patients treated at our safety-net hospital had higher odds of enrollment than those treated at our university hospital (OR: 2.62, 95% CI: 1.11-5.99, P = 0.02). CONCLUSIONS Efforts to improve diversity in biomedical research cannot consider minority patients as a monolithic cohort. Instead, targeted interventions that address diverse cultural concerns and improve access to enrollment at safety-net centers are requisite.
Collapse
Affiliation(s)
- Morgan F Pettigrew
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Amr I Al Abbas
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Anida Southichack
- Tissue Management Shared Resource, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michelle R Ju
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Suntrea T G Hammer
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yulun Liu
- Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, Texas
| | - Matthew R Porembka
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sam C Wang
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
31
|
Li C, Xu T, Hou G, Wang Y, Fu Q. DNA nanotechnology-based strategies for gastric cancer diagnosis and therapy. Mater Today Bio 2025; 30:101459. [PMID: 39866794 PMCID: PMC11762204 DOI: 10.1016/j.mtbio.2025.101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Gastric cancer (GC) is a formidable adversary in the field of oncology. The low early diagnosis rate of GC results in a low overall survival rate. Therefore, early accurate diagnosis and effective treatment are the key to reduce the mortality of GC. With the advent of nanotechnology, researchers continue to explore new possibilities for accurate diagnosis and effective treatment. One such breakthrough is the application of DNA nanotechnology. In this paper, the application of exciting DNA nanomaterials in the diagnosis and treatment of GC is discussed in depth. Firstly, the biomarkers related to GC and the diagnostic strategies related to DNA nanotechnology are summarized. Second, the latest research progress of DNA nanomaterials in the GC targeted therapy are summarized. Finally, the challenges and opportunities of DNA nanomaterials in the research and clinical application of GC are prospected.
Collapse
Affiliation(s)
- Congcong Li
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Tongyang Xu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Guopeng Hou
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Yin Wang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Qinrui Fu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| |
Collapse
|
32
|
Huamani Ortiz ADJ, Campos Segura AV, Magaño Bocanegra KJ, Velásquez Sotomayor MB, Barrón Pastor HJ, Llimpe Mitma de Barrón Y, Chacón Villanueva RD, Murillo Carrasco AG, Ortiz Rojas CA. Transcriptome-Based Survival Analysis Identifies MAP4K4 as a Prognostic Marker in Gastric Cancer with Microsatellite Instability. Cancers (Basel) 2025; 17:412. [PMID: 39941781 PMCID: PMC11816344 DOI: 10.3390/cancers17030412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Gastric cancer (GC) is a highly aggressive malignancy with diverse molecular subtypes. While microsatellite instability (MSI) GC generally carries a favorable prognosis, a subset of patients experiences poor outcomes, highlighting the need for refined prognostic markers. Methods: This study utilized transcriptomic and clinical data from two independent cohorts, The Cancer Genome Atlas (TCGA) and the Asian Cancer Research Group (ACRG), to identify novel prognostic genes in MSI-GC. Results: Through rigorous survival analysis, we identified high MAP4K4 expression (MAP4K4high) as an independent and robust predictor of poor overall survival (OS) and disease-free survival (DFS) specifically within the MSI-GC subtype. MAP4K4high was associated with increased hazard ratios for both OS and DFS in both cohorts, even after adjusting for clinicopathological factors. Further analysis revealed that MAP4K4high MSI-GC tumors exhibit a distinct molecular profile characterized by increased extracellular matrix remodeling, epithelial-mesenchymal transition, and a microenvironment enriched in monocytes and cancer-associated fibroblasts (CAFs). Notably, a subgroup of MSI-GC patients with a CIN-like phenotype and high MAP4K4 expression exhibited particularly dismal outcomes. Conclusions: Our findings establish MAP4K4 as a promising prognostic biomarker for risk stratification in MSI-GC and suggest its potential role in driving aggressive tumor behavior through modulation of the tumor microenvironment.
Collapse
Affiliation(s)
- Alvaro De Jesus Huamani Ortiz
- Molecular Medicine Research and Teaching Group (MEDMOL), Faculty of Medicine, National University of San Marcos, Lima 15081, Peru; (A.D.J.H.O.); (H.J.B.P.); (Y.L.M.d.B.)
- Immunology and Cancer Research Group (IMMUCA), OMICS, Lima 15001, Peru; (A.V.C.S.); (M.B.V.S.); (R.D.C.V.)
| | - Anthony Vladimir Campos Segura
- Immunology and Cancer Research Group (IMMUCA), OMICS, Lima 15001, Peru; (A.V.C.S.); (M.B.V.S.); (R.D.C.V.)
- Clinical and Functional Genomics Group, International Center of Research CIPE, A.C. Camargo Cancer Center, Sao Paulo 01509-010, Brazil
| | - Kevin Jorge Magaño Bocanegra
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | - Mariana Belén Velásquez Sotomayor
- Immunology and Cancer Research Group (IMMUCA), OMICS, Lima 15001, Peru; (A.V.C.S.); (M.B.V.S.); (R.D.C.V.)
- Faculty of Medicine, Southern Scientific University, Lima 150142, Peru
| | - Heli Jaime Barrón Pastor
- Molecular Medicine Research and Teaching Group (MEDMOL), Faculty of Medicine, National University of San Marcos, Lima 15081, Peru; (A.D.J.H.O.); (H.J.B.P.); (Y.L.M.d.B.)
| | - Yesica Llimpe Mitma de Barrón
- Molecular Medicine Research and Teaching Group (MEDMOL), Faculty of Medicine, National University of San Marcos, Lima 15081, Peru; (A.D.J.H.O.); (H.J.B.P.); (Y.L.M.d.B.)
| | - Ruy Diego Chacón Villanueva
- Immunology and Cancer Research Group (IMMUCA), OMICS, Lima 15001, Peru; (A.V.C.S.); (M.B.V.S.); (R.D.C.V.)
- Department of Pathology, School of Veterinary Medicine, University of São Paulo, São Paulo 05508-900, Brazil
| | - Alexis Germán Murillo Carrasco
- Immunology and Cancer Research Group (IMMUCA), OMICS, Lima 15001, Peru; (A.V.C.S.); (M.B.V.S.); (R.D.C.V.)
- Center for Translational Research in Oncology (LIM/24), Hospital das Clínicas, Faculty of Medicine, University of São Paulo (HCFMUSP), São Paulo 01246-000, Brazil
- Comprehensive Center for Precision Oncology, University of São Paulo, São Paulo 01246-000, Brazil
| | - César Alexander Ortiz Rojas
- Immunology and Cancer Research Group (IMMUCA), OMICS, Lima 15001, Peru; (A.V.C.S.); (M.B.V.S.); (R.D.C.V.)
- Center for Translational Research in Oncology (LIM/24), Hospital das Clínicas, Faculty of Medicine, University of São Paulo (HCFMUSP), São Paulo 01246-000, Brazil
- Medical Investigation Laboratory in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM/31), Hospital das Clínicas, Faculty of Medicine, University of São Paulo (HCFMUSP), São Paulo 01246-000, Brazil
| |
Collapse
|
33
|
Panahizadeh R, Panahi P, Asghariazar V, Makaremi S, Noorkhajavi G, Safarzadeh E. A literature review of recent advances in gastric cancer treatment: exploring the cross-talk between targeted therapies. Cancer Cell Int 2025; 25:23. [PMID: 39856676 PMCID: PMC11762578 DOI: 10.1186/s12935-025-03655-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) ranks fourth in global mortality rates and fifth in prevalence, making it one of the most common cancers worldwide. Recent clinical studies have highlighted the potential of immunotherapies as a promising approach to treating GC. This study aims to shed light on the most impactful therapeutic strategies in the context of GC immunotherapy, highlighting both established and emerging approaches. MAIN BODY This review examines over 160 clinical studies conducted globally, focusing on the effectiveness of various immunotherapy modalities, including cancer vaccines, adoptive cell therapy, immune checkpoint inhibitors (ICIs), and monoclonal antibodies (mAbs). A comprehensive search of peer-reviewed literature was performed using databases such as Web of Science, PubMed, and Scopus. The selection criteria included peer-reviewed articles published primarily within the last 10 years, with a focus on studies that provided insights into targeted therapies and their mechanisms of action, clinical efficacy, and safety profiles. The findings indicate that these immunotherapy strategies can enhance treatment outcomes for GC, aligning with current treatment guidelines. ICIs like pembrolizumab and nivolumab have shown significant survival benefits in specific GC subgroups. Cancer vaccines and CAR-T cell therapies demonstrate potential, while mAbs targeting HER2 and VEGFR pathways enhance outcomes in combination regimens. We discuss the latest advancements and challenges in targeted therapy and immunotherapy for GC. Given the evolving nature of this field, this research emphasizes significant evidence-based therapies and those currently under evaluation rather than providing an exhaustive overview. Challenges include resistance mechanisms, immunosuppressive tumor environments, and inconsistent results from combination therapies. Biomarker-driven approaches and further research into emerging modalities like CAR-T cells and cancer vaccines are critical for optimizing treatments. CONCLUSIONS Immunotherapy is reshaping GC management by improving survival and quality of life. Ongoing research and clinical evaluations are crucial for refining personalized and effective therapies.
Collapse
Affiliation(s)
- Reza Panahizadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Padideh Panahi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shima Makaremi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ghasem Noorkhajavi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 85991-56189, Iran.
| |
Collapse
|
34
|
Guo X, Liu Y. Capecitabine enhances sensitivity to oxaliplatin in advanced gastric cancer and the effects on patients' FOXP1 and GGT levels. BMC Gastroenterol 2025; 25:35. [PMID: 39856546 PMCID: PMC11759413 DOI: 10.1186/s12876-025-03624-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE To investigate the effect of capecitabine on the sensitivity of oxaliplatin and on the level of transcription factor forkhead box P1 (FOXP1) and gamma-glutamyl transpeptidase (GGT) in patients with intermediate and advanced gastric cancer. METHODS A total of 152 Patients with advanced gastric cancer who were continuously diagnosed and treated in our hospital were selected as the study objects. The general data were retrospectively analyzed. The patients in the control group received oxaliplatin, while the patients in the study group received capecitabine on the basis of the control group. The FOXP1 expression level was detected using immunohistochemistry. Serum levels of GGT were measured by chemiluminescence. Protein levels were detected by Western blot. The prognostic factors were analyzed by the COX regression model. The Kaplan-Meier survival curve was used to analyze the survival of gastric cancer. RESULTS The effective rates (complete response, partial response, and stability) of the study group and the control group were 94.74% and 76.32%, respectively. Compared with adjacent normal tissues, the expression level of FOXP1 in gastric cancer tissues was lower (P < 0.05). After treatment, the average expression level of FOXP1 in the gastric cancer tissue of the study group was higher than the control group (P < 0.05). Moreover, lower FOXP1 expression was associated with lower overall survival (OS) (1-year survival and 3-year survival were 75.76% and 53.03%, respectively) (P < 0.05). Further analysis showed that capecitabine combined with oxaliplatin down-regulated the expression of DNA repair related-proteins and up-regulated the expression of key molecules of the apoptosis pathway, thus enhancing the killing effect of oxaliplatin on gastric cancer cells (P < 0.05). Both the 1-year and 3-year survival rates of the study group were higher than that in the control group (P < 0.05). The 1-year survival rate of 152 patients with gastric cancer was 84.87% (129/152) and the 3-year survival rate was 63.17% (96/152). Age, tumor-node-metastasis (TNM) stage, lymph node metastasis, chemotherapy regimen, FOXP1, and GGT levels were important factors in determining OS. CONCLUSION Capecitabine effectively enhanced the sensitivity of intermediate and advanced gastric cancer to oxaliplatin, improved the therapeutic effect and ameliorated the prognosis of patients.
Collapse
Affiliation(s)
- Xinyu Guo
- Department of General Surgery, Fuwai Central China Cardiovascular Hospital, No. 1, Fuwai Road, Zhengdong New District, Zhengzhou, Henan, 451460, PR China.
| | - Yi Liu
- Department of General Surgery, Fuwai Central China Cardiovascular Hospital, No. 1, Fuwai Road, Zhengdong New District, Zhengzhou, Henan, 451460, PR China
| |
Collapse
|
35
|
Wang R, Liu G, Wang K, Pan Z, Pei Z, Hu X. Hypoxia signature derived from tumor-associated endothelial cells predict prognosis in gastric cancer. Front Cell Dev Biol 2025; 13:1515681. [PMID: 39901877 PMCID: PMC11788339 DOI: 10.3389/fcell.2025.1515681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/03/2025] [Indexed: 02/05/2025] Open
Abstract
Background A hypoxic metabolism environment in the tumors is often associated with poor prognostic events such as tumor progression and treatment resistance. In gastric cancer, the mechanism of how hypoxia metabolism affects the tumor microenvironment and immunotherapy efficacy remains to be elucidated. Methods We used the bulk-mapping method to analyze the signatures correlated with the response of immunotherapy in the single-cell dataset. Cellular, pathway, and gene were systematically analyzed in both single-cell and bulk validation datasets. Results The most significant cell proportion difference between the response and non-response groups was in endothelial cells, which represent the malignant cells. VWF was specifically overexpressed in endothelial cells and was the hub gene of differential genes. EPAS1 was a VWF trans-regulated gene and highly positively correlated with VWF in expression. Knockdown experiments demonstrated that siVWF reduced the expression of VWF, EPAS1, and HIF1A, as well as the synthesis of lactate and adenosine which are indicators of hypoxic metabolism. These results suggest that the overexpression of core malign endothelial genes such as VWF drives hypoxic metabolism in tumors and creates an immunosuppressive environment that reduces the efficacy of immunotherapy. The adverse prognosis of the hypoxia signature was validated in the bulk cohort and significance was further enhanced after selecting core genes and combined survival weight scoring. Conclusion In summary, high expression of the malignant endothelial cell driver genes VWF and EPAS1 enhances hypoxic metabolism, and malignant cell-immune cell interactions suppress the immune response. Therefore, the two core genes of hypoxic metabolism might represent potential therapeutic and predicting biomarkers for immunotherapy of gastric cancer in the future.
Collapse
Affiliation(s)
- Ruiheng Wang
- Surgical Ward, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Guijun Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of administrative, The Fourth Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ke Wang
- Endoscopy Room, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Zhanglei Pan
- Surgical Ward, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhihua Pei
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Xijiao Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Postdoctoral Research Station of Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
36
|
Mazurek M, Jaros M, Gliwa AM, Sitarz MZ, Dudzińska E, Zinkiewicz K, Sitarz R. Epstein-Barr Virus (EBV) and Human Papilloma Virus (HPV) in Gastric Cancers, with Special Reference to Gastric Cancer at a Young Age-A Pilot Study in Poland. Int J Mol Sci 2025; 26:711. [PMID: 39859425 PMCID: PMC11765604 DOI: 10.3390/ijms26020711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Gastric cancer (GC) is one of the most common cancers in the world. It is a multi-factorial disease influenced by both genetic and environmental factors such as diet, obesity, radiation exposure, and infectious agents. Viral infections usually lead to chronic inflammation, which can initiate the development of cancers. To date, only a few studies have been published about Epstein-Barr virus (EBV) and human papillomavirus (HPV) infections in the context of the development of GC. In particular, research on the development of cancer among people under 45 years of age, including the impacts of EBV and HPV, is rare, and clear results have not been obtained. The aim of this study was to analyze the frequency of occurrence of EBV and HPV in GC, particularly in early-onset gastric cancer (EOGC). Tissue material from 135 patients with GC, including 84 men and 51 women, was examined. RT-PCR was performed to detect EBV, and PCR was performed to detect HPV. There were no significant impacts of EBV and HPV infections on any subtype of GC. There was also no statistically significant dependence of gender and location of the tumor on any subtype of GC. Further research on the impacts of infectious agents such as EBV and HPV on GC should be conducted using larger populations.
Collapse
Affiliation(s)
- Marek Mazurek
- Department of Surgical Oncology, Masovian Cancer Hospital, 05-135 Wieliszew, Poland;
- Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-950 Lublin, Poland; (M.J.); (A.M.G.)
| | - Małgorzata Jaros
- Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-950 Lublin, Poland; (M.J.); (A.M.G.)
| | - Anna M. Gliwa
- Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-950 Lublin, Poland; (M.J.); (A.M.G.)
| | - Monika Z. Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Ewa Dudzińska
- Department of Dietetics and Nutrition Education, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Krzysztof Zinkiewicz
- Independent Laboratory of Diagnostic, Interventional Endoscopy of the Department of Oncology, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Robert Sitarz
- Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-950 Lublin, Poland; (M.J.); (A.M.G.)
- Department of Surgical Oncology, St. John’s Cancer Center, 20-090 Lublin, Poland
| |
Collapse
|
37
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
38
|
Shi B, Wang W, Fang S, Wu S, Zhu L, Chen Y, Dong H, Yan F, Yuan F, Ye J, Zhang H, Lin LL. Raman spectroscopy analysis combined with computed tomography imaging to identify microsatellite instability in gastric cancers. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 325:125062. [PMID: 39226670 DOI: 10.1016/j.saa.2024.125062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/05/2024] [Accepted: 08/25/2024] [Indexed: 09/05/2024]
Abstract
Accurate determination of microsatellite instability (MSI) status is critical for tailoring treatment approaches for gastric cancer patients. Existing clinical techniques for MSI diagnosis are plagued by problems of suboptimal time efficiency, high cost, and burdensome experimental requirements. Here, we for the first time establish the classification model of gastric cancer MSI status based on Raman spectroscopy. To begin with, we reveal that tumor heterogeneity-induced signal variations pose a prominent impact on MSI classification. To eliminate this issue, we develop Euclidean distance-based Raman Spectroscopy (EDRS) algorithm, which establishes a standard spectrum to represent the "most microsatellite stable" status. The similarity between each spectrum of tissues with the standard spectrum is calculated to provide a direct assessment on the MSI status. Compared to machine learning-algorithms including k-Nearest Neighbors, Random Forest, and Extreme Learning Machine, the EDRS method shows the highest accuracy of 94.6 %. Finally, we integrate the EDRS method with the clinical diagnostic modality, computed tomography, to construct an innovative joint classification model with good classification performance (AUC = 0.914, Accuracy = 94.6 %). Our work demonstrates a robust, rapid, non-invasive, and convenient tool in identifying the MSI status, and opens new avenues for Raman techniques to fit into existing clinical workflow.
Collapse
Affiliation(s)
- Bowen Shi
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Wenfang Wang
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, PR China
| | - Shiyan Fang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Siyi Wu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Lan Zhu
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Yong Chen
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Haipeng Dong
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Jian Ye
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China; Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China.
| | - Linley Li Lin
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| |
Collapse
|
39
|
Wang Y, Li X, Wang Y, Qin J. Establishment and characterization of a new mouse gastric carcinoma cell line, MCC. Cancer Cell Int 2025; 25:9. [PMID: 39800685 PMCID: PMC11727671 DOI: 10.1186/s12935-024-03633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The aim of this study was to establish a primary mouse gastric carcinoma cell line. METHODS Gastric adenocarcinoma in the body region was induced in immunocompetent BALB/c mice using N-Methyl-N-nitrosourea and a 2% NaCl solution. Fresh gastric cancer tissue samples were cultured in 1640 medium supplemented with 10% fetal bovine serum for primary culture and subculture. Cellular morphology was assessed via light microscopy, and a cell growth curve was established. Genomic and proteomic analyses were conducted to characterize the molecular features of the cell lines. This cell line demonstrated a 100% success rate in forming subcutaneous tumors in BALB/c mice. By integrating proteomic profiles from clinical gastric cancer patients and the murine subcutaneous tumor model, several molecular targets suitable for preclinical investigation were identified. Trametinib, a MEK inhibitor, was employed as a model compound in our preclinical study. RESULTS A novel gastric carcinoma cell line, designated MCC, was established from BALB/c mice. This cell line exhibited a doubling time of approximately 33 h. Genomic and proteomic analyses identified mutations frequently observed in clinical gastric cancer patients, such as Kras, Egfr, and Ccnd3. Additionally, MCC overexpresses proteins, including SLC1A5, MCM6, and ITGA2, which are significantly upregulated in gastric cancer tissues compared to adjacent non-cancerous tissues. The MCC cell line demonstrated stable tumorigenicity in immunocompetent BALB/c mice, forming subcutaneous tumors that closely resemble the proteomic profile of clinical gastric cancer samples. This high concordance facilitated the identification of several potential therapeutic targets for gastric cancer. Preclinical studies with trametinib revealed that treatment effectively inhibited gastric cancer growth, likely mediated through the activation of immune cells, particularly neutrophils and T cells. CONCLUSIONS The MCC cell line serves as an indispensable model for gastric cancer research, offering a robust platform for investigating tumor development and progression. Its exceptional tumorigenic capacity and strong concordance with clinical proteomic profiles underscore its significance in translational research, facilitating the discovery of novel therapeutic targets and elucidation of molecular pathways critical for developing effective treatment strategies.
Collapse
Affiliation(s)
- Yushen Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, People's Republic of China
| | - Xianju Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, People's Republic of China.
| | - Yi Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, People's Republic of China.
| | - Jun Qin
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, People's Republic of China.
| |
Collapse
|
40
|
Xiao Y, Zhu J, Xie H, Wang Z, Huang Z, Su M. Intratumoral and peritumoral radiomics for forecasting microsatellite status in gastric cancer: a multicenter study. BMC Cancer 2025; 25:66. [PMID: 39794732 PMCID: PMC11724602 DOI: 10.1186/s12885-025-13450-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
OBJECTIVE This investigation attempted to examine the effectiveness of CT-derived peritumoral and intratumoral radiomics in forecasting microsatellite instability (MSI) status preoperatively among gastric cancer (GC) patients. METHODS A retrospective analysis was performed on GC patients from February 2019 to December 2023 across three healthcare institutions. 364 patients (including 41 microsatellite instability-high (MSI-H) and 323 microsatellite instability-low/stable (MSI-L/S)) were stratified into a training set (n = 202), an internal validation set (n = 84), and an external validation set (n = 78). Radiomics features were obtained from both the intratumoral region (IR) and the intratumoral plus 3-mm peritumoral region (IPR) on preoperative contrast-enhanced CT images. After standardizing and reducing the dimensionality of these features, six radiomic models were constructed utilizing three machine learning techniques: Support Vector Machine (SVM), Linear Support Vector Classification (LinearSVC), and Logistic Regression (LR). The optimal model was determined by evaluating the Receiver Operating Characteristic (ROC) curve's Area Under the Curve (AUC), and the radiomics score (Radscore) was computed. A clinical model was developed using clinical characteristics and CT semantic features, with the Radscore integrated to create a combined model. Used ROC curves, calibration plots, and Decision Curve Analysis (DCA) to assess the performance of radiomics, clinical, and combined models. RESULTS The LinearSVC model using the IPR achieved the highest AUC of 0.802 in the external validation set. The combined model yielded superior AUCs in internal and external validation sets (0.891 and 0.856) in comparison to clinical model [(0.724, P = 0.193) and (0.655, P = 0.072)] and radiomics model [(0.826, P = 0.160) and (0.802, P = 0.068)]. Furthermore, results from calibration and DCA underscored the model's suitability and clinical relevance. CONCLUSION The combined model, which integrates IPR radiomics with clinical characteristics, accurately predicts MSI status and supports the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Yunzhou Xiao
- Department of Radiology, The People's Hospital of PingYang, Wenzhou Medical University, Wenzhou, 325400, China
| | - Jianping Zhu
- Department of Radiology, Ningbo Yinzhou NO.2 Hospital, Ningbo, 315100, China
| | - Huanhuan Xie
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Zhongchu Wang
- Department of Radiology, The People's Hospital of PingYang, Wenzhou Medical University, Wenzhou, 325400, China
| | - Zhaohai Huang
- Department of Radiology, The People's Hospital of PingYang, Wenzhou Medical University, Wenzhou, 325400, China.
| | - Miaoguang Su
- Department of Radiology, The People's Hospital of PingYang, Wenzhou Medical University, Wenzhou, 325400, China.
| |
Collapse
|
41
|
Carbone L, Incognito GG, Incognito D, Nibid L, Caruso G, Berretta M, Taffon C, Palumbo M, Perrone G, Roviello F, Marrelli D. Clinical implications of epithelial-to-mesenchymal transition in cancers which potentially spread to peritoneum. Clin Transl Oncol 2025:10.1007/s12094-024-03837-2. [PMID: 39775727 DOI: 10.1007/s12094-024-03837-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a biological process by which epithelial cells increase their motility and acquire invasive capacity. It represents a crucial driver of cancer metastasis and peritoneal dissemination. EMT plasticity, with cells exhibiting hybrid epithelial/mesenchymal states, and its reverse process, mesenchymal-to-epithelial transition (MET), allows them to adapt to different microenvironments and evade therapeutic intervention. Resistance to conventional treatments, including chemotherapy, is a major problem. Therapies targeting EMT may inhibit tumour cell migration and invasion, while affecting normal cells and repair mechanisms, resulting in potential side effects. This paper addresses the question of the impact of EMT status on cancers with potential spread to the peritoneum, which has remained unclear in literature. Relevant studies were selected from 2000 to 2024. Three macrosections were analysed: (i) pathological characteristics, (ii) surgical implications and (iii) oncological therapies. The focus was on survival and peritoneal recurrence time in patients who underwent surgical treatment.
Collapse
Affiliation(s)
- Ludovico Carbone
- Unit of Surgical Oncology, Department of Medicine Surgery and Neuroscience, University of Siena, Viale Mario Bracci 16, 53100, Siena, Italy.
| | - Giosuè Giordano Incognito
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123, Catania, Italy
| | - Dalila Incognito
- Department of Human Pathology "G. Barresi", School of Specialization in Medical Oncology Unit, University of Messina, 98122, Messina, Italy
| | - Lorenzo Nibid
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, 00128, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128, Roma, Italy
| | - Giuseppe Caruso
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123, Catania, Italy
| | - Massimiliano Berretta
- Department of Human Pathology "G. Barresi", School of Specialization in Medical Oncology Unit, University of Messina, 98122, Messina, Italy
- Department of Clinical and Experimental Medicine, University of Messina, 98122, Messina, Italy
| | - Chiara Taffon
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, 00128, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128, Roma, Italy
| | - Marco Palumbo
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123, Catania, Italy
| | - Giuseppe Perrone
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, 00128, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128, Roma, Italy
| | - Franco Roviello
- Unit of Surgical Oncology, Department of Medicine Surgery and Neuroscience, University of Siena, Viale Mario Bracci 16, 53100, Siena, Italy
| | - Daniele Marrelli
- Unit of Surgical Oncology, Department of Medicine Surgery and Neuroscience, University of Siena, Viale Mario Bracci 16, 53100, Siena, Italy
| |
Collapse
|
42
|
Yu X, Shao Y, Dong H, Yan J, Zhang X, Ye G. Molecular subtype of gastric cancer based on apoptosis-related genes reveals differential immune microenvironment and intratumoral microorganisms distribution. BMC Cancer 2025; 25:12. [PMID: 39762768 PMCID: PMC11702164 DOI: 10.1186/s12885-024-13411-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is known for its high heterogeneity, presenting challenges in current clinical treatment strategies. Accurate subtyping and in-depth analysis of the molecular heterogeneity of GC at the molecular level are still not fully understood. METHODS This study categorized GC into two subtypes based on apoptosis-related genes (ARGs) and investigated differences in tumor immune microenvironment, intratumoral microorganisms distribution, gene expression, and signaling pathways. Key prognostic genes related to apoptosis in GC were identified through random survival forest analysis, and their specific signaling mechanisms were explored. Expression levels of key genes were validated through PCR in paired GC tissues and cancer cell lines. Moreover, biological functions of these key genes were verified in vitro experiments. RESULTS A consistent clustering of GC was conducted using 161 apoptosis-related genes (ARGs), resulting in the identification of two subtypes, C1 and C2. Subsequently, significant differences were found in the tumor immune microenvironment, intratumoral microorganisms, gene expression, signaling pathways, and protein interaction networks between the two subtypes. GPX3, PLAT, and CAV1 were identified as key prognostic genes related to apoptosis in GC, with a focus on their impact on disease progression-related pathways. Furthermore, PCR assays validated that these three key genes exhibited significantly low expression levels in both GC cell lines and tissues. Finally, knocking down key genes expression significantly promoted cell proliferation, colony formation and invasion of GC. CONCLUSIONS Our study conducted a comprehensive analysis of the molecular characteristics of ARGs in GC, revealed their association with the tumor immune microenvironment and intratumoral microorganisms. These findings provide new ideas for the molecular classification of GC.
Collapse
Affiliation(s)
- Xuan Yu
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| | - Yongfu Shao
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, China.
- Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Haotian Dong
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jianing Yan
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| | - Xinjun Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| | - Guoliang Ye
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| |
Collapse
|
43
|
Wu W, Tao G, Chen Z, Zhou Q. Identification of disulfidptosis-related subtypes in gastric cancer and GAMT is a key gene during disulfidptosis. Sci Rep 2025; 15:111. [PMID: 39747986 PMCID: PMC11697009 DOI: 10.1038/s41598-024-83580-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Recent studies have found that disulfidptosis occurs in cells under glucose starvation. The role of this programmed death method in gastric cancer remains to be explored. Cluster analysis based on disulfidptosis related genes to analyze the differential characteristics of disulfidptosis subtypes. We construct a prognostic risk model using 12 differentially expressed genes of disulfidptosis subtypes. We also analyzed the disulfidptosis subtypes at single-cell resolution. We found that cluster 1 has a poor prognosis and is characterized by a younger age. Inhibiting the expression of GAMT genes associated with disulfidptosis subtypes can significantly inhibit the proliferation of gastric cancer cells, which may be an important target for gastric cancer treatment. Cluster 2 patients are more sensitive to various chemotherapy drugs and immunotherapy. Mesenchymal cells, especially myCAF, endothelial cells, and smooth muscle cells, have strong disulfidptosis scores. In summary, our study provides new insights into the role of disulfidptosis in gastric cancer, and this may be used to guide the treatment of gastric cancer.
Collapse
Affiliation(s)
- Wenxue Wu
- Department of General Surgery, Shanghai Punan Hospital, No.279 Linyi Road, Pudong New District, Shanghai, China
| | - Guoqiang Tao
- Department of General Surgery, Shanghai Punan Hospital, No.279 Linyi Road, Pudong New District, Shanghai, China
| | - Zhiqing Chen
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi medical college, Nanchang University, Jiangxi, China
| | - Qi Zhou
- Department of General Surgery, Shanghai Punan Hospital, No.279 Linyi Road, Pudong New District, Shanghai, China.
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi medical college, Nanchang University, Jiangxi, China.
| |
Collapse
|
44
|
Tian Z, Yang L, Yang R, Yang W. The prognostic and immunomodulatory role of the MMR system in patients with stomach adenocarcinoma. Sci Rep 2025; 15:180. [PMID: 39748125 PMCID: PMC11695722 DOI: 10.1038/s41598-024-84613-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025] Open
Abstract
The mismatch repair (MMR) system plays a crucial role in the maintenance of DNA replication fidelity and genomic stability. The clinical value of the MMR molecular marker as an immunotherapy for advanced solid tumors has been documented. However, this therapy is not effective in some patients. This study aimed to develop an MMR-related molecular prognostic model for identifying appropriate populations of stomach adenocarcinoma (STAD) for better treatment outcome. The MMR genes expression data were downloaded from TCGA and CCLE databases. The expression of four MMR genes, construction of a prognostic risk model, and assessment of immune infiltration in STAD were performed using Xiantao online tool. GEPIA2 was used to explore the association of MMR genes expression with clinical stage and overall survival. The frequency and prognostic value of MMR genes in STAD were conducted on the cBioPortal. The MLH1 co-expression network was established based on the LinkedOmics database. This study found that the expression of MSH2, MSH6 and PMS2 was up-regulated in STAD tissues. Moreover, differential MMR genetic expression levels were not significantly correlated with the clinical stages of STAD. Besides, no significant difference in PFS or OS was observed in STAD patients with or without MMR genetic alteration. Moreover, MLH1 and MSH2 were used to establish a prognostic risk model. The immune infiltration levels of most immune cells were upregulated in the high-risk group with elevated expression of PDCD1 and low TMB score. Finally, we found that MLH1 was an independent predictor of STAD prognosis among the four MMR genes. An MMR-related prognostic model for STAD was constructed based on genes. This model provides a new therapeutic concept for the diagnosis and treatment of STAD.
Collapse
Affiliation(s)
- Zhihui Tian
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan City, 030013, Shanxi Province, China.
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital, Shanxi Medical University, No.3, Staff New Street, Xinghualing District, Taiyuan City, 030000, Shanxi Province, China.
| | - Lili Yang
- Department of Intensive Care Unit, Shanxi Hospital Affiliated to Cancer Hospital, Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Shanxi Medical University, Taiyuan City, 030013, Shanxi Province, China
| | - Rong Yang
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan City, 030013, Shanxi Province, China.
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital, Shanxi Medical University, No.3, Staff New Street, Xinghualing District, Taiyuan City, 030000, Shanxi Province, China.
| | - Wenhui Yang
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan City, 030013, Shanxi Province, China.
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital, Shanxi Medical University, No.3, Staff New Street, Xinghualing District, Taiyuan City, 030000, Shanxi Province, China.
| |
Collapse
|
45
|
Laurino S, Russi S, Sabato C, Luongo M, Laurenziello P, Vagliasindi A, Di Stefano G, Vita GAC, Patitucci G, Amendola E, Zoppoli P, Albano F, Balzamo C, Notarangelo T, Falco G. The inhibition of SLC8A1 promotes Ca 2+-dependent cell death in Gastric Cancer. Biomed Pharmacother 2025; 182:117787. [PMID: 39731939 DOI: 10.1016/j.biopha.2024.117787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
Intracellular Ca2+ homeostasis dysregulation, through the modulation of calcium permeable ion channels and transporters, is gaining attention in cancer research as an apoptosis evasion mechanism. Recently, we highlighted a prognostic role for several calcium permeable channels. Among them, here, we focused on the plasma membrane bidirectional Na+/Ca2+ exchanger SLC8A1. Data from Kaplan-Meier plotter and The Cancer Genome Atlas were used to evaluate in silico the association of SLC8A1 expression with Gastric Cancer (GC) patients' survival, and its levels in different patient subgroups. In vitro experiments were used to explore SLC8A1 as a possible target in GC. Interestingly, SLC8A1 expression was associated with a worst prognosis, and resulted up-regulated in diffuse/poorly-cohesive histological GC type, Genomically Stable samples and in advanced TNM stages. We demonstrated that SLC8A1 selective pharmacological inhibition, through CB-DMB, significantly reduced cancer proliferation and induced Ca2+-dependent cell death in GC cells, both alone and synergically with cisplatin treatment. SLC8A1 inhibition could represents a potential subgroup-specific therapeutic approach for GC patients based on its ability to induce Ca2+-dependent cell death.
Collapse
Affiliation(s)
- Simona Laurino
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy
| | - Sabino Russi
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy.
| | - Claudia Sabato
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy
| | - Margherita Luongo
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy
| | - Pasqualina Laurenziello
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy
| | - Alessio Vagliasindi
- Unit of Abdominal Oncological Surgery, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy
| | - Greta Di Stefano
- Unit of Abdominal Oncological Surgery, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy
| | - Giulia Anna Carmen Vita
- Anatomical Pathology Department, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy
| | - Giuseppe Patitucci
- Anatomical Pathology Department, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy
| | - Elena Amendola
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council (CNR), Naples, Italy
| | - Pietro Zoppoli
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Francesco Albano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Chiara Balzamo
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Tiziana Notarangelo
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, PZ, Italy
| | - Geppino Falco
- Department of Biology, University of Naples Federico II, Naples, Italy; Biogem, Istituto di Biologia e Genetica Molecolare, Ariano Irpino, AV, Italy
| |
Collapse
|
46
|
da Silva EL, Mesquita FP, Pinto LC, Gomes BPS, de Oliveira EHC, Burbano RMR, Moraes MEAD, de Souza PFN, Montenegro RC. Transcriptome analysis displays new molecular insights into the mechanisms of action of Mebendazole in gastric cancer cells. Comput Biol Med 2025; 184:109415. [PMID: 39566281 DOI: 10.1016/j.compbiomed.2024.109415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/15/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
Gastric cancer (GC) is a common cancer worldwide. Therefore, searching for effective treatments is essential, and drug repositioning can be a promising strategy to find new potential drugs for GC therapy. For the first time, we sought to identify molecular alterations and validate new mechanisms related to Mebendazole (MBZ) treatment in GC cells through transcriptome analysis using microarray technology. Data revealed 1066 differentially expressed genes (DEGs), of which 345 (2.41 %) genes were upregulated, 721 (5.04 %) genes were downregulated, and 13,231 (92.54 %) genes remained unaltered after MBZ exposure. The overexpressed genes identified were CCL2, IL1A, and CDKN1A. In contrast, the H3C7, H3C11, and H1-5 were the top 3 underexpressed genes. Gene set enrichment analysis (GSEA) identified 8 pathways significantly overexpressed in the treated group (p < 0.05 and FDR<0.25). The validation of the expression of top desregulated genes by RT-qPCR confirmed the transcriptome results, where MBZ increased the CCL2, IL1A, and CDKN1A and reduced the H3C7, H3C11, and H1-5 transcript levels. Expression analysis in samples from TCGA databases correlated that the lower ILI1A and higher H3C11 and H1-5 gene expression are associated with decreased overall survival rates in patients with GC, indicating that MBZ treatment can improve the prognosis of patients. Thus, the data demonstrated that the drug MBZ alters the transcriptome of the AGP-01 lineage, mainly modulating the expression of histone proteins and inflammatory cytokines, indicating a possible epigenetic and immunological effect on tumor cells, these findings highlight new mechanisms of action related to MBZ treatment. Additional studies are still needed to better clarify the epigenetic and immune mechanism of MBZ in the therapy of GC.
Collapse
Affiliation(s)
- Emerson Lucena da Silva
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil
| | - Felipe Pantoja Mesquita
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil
| | - Laine Celestino Pinto
- Laboratory of Experimental Neuropathology, Biological Science Institute, Federal University of Pará, Mundurucus Street, Belém, Brazil
| | - Bruna Puty Silva Gomes
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SAMAM), Evandro Chagas Institute (IEC), Ananindeua, Brazil
| | | | - Rommel Mario Rodríguez Burbano
- Molecular Biology Laboratory, Ophir Loyola Hospital, Av. Governador Magalhães Barata, Belém, Brazil; Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Augusto Correa Avenue, Belém, Brazil
| | - Maria Elisabete Amaral de Moraes
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil
| | - Pedro Filho Noronha de Souza
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil; Visiting Researcher at the Cearense Foundation to Support Scientific and Technological Development, Brazil; National Institute of Science and Technology in Human Pathogenic Fungi, Ribeirão Preto, Brazil.
| | - Raquel Carvalho Montenegro
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil; Red Latinoamericana de Implementación y Validación de guias clinicas Farmacogenomicas (RELIVAF), Brazil.
| |
Collapse
|
47
|
Yoshioka K, Nakano Y, Horichi M, Aono D, Takeshita Y, Takamura T. Metastatic Pheochromocytoma/Paraganglioma Overproducing Multiple Catecholamines. JCEM CASE REPORTS 2025; 3:luae241. [PMID: 39726667 PMCID: PMC11669864 DOI: 10.1210/jcemcr/luae241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Indexed: 12/28/2024]
Abstract
Pheochromocytoma and paraganglioma (PPGL) are rare chromaffin-cell tumors producing adrenaline and/or noradrenaline, or solely dopamine. A 52-year-old man presenting with hypertension (141/79 mm Hg) and weight loss (10 kg in 6 months) was admitted to our hospital. Computed tomography revealed a massive right adrenal mass (150 mm) with partial necrosis, accompanied by multiple liver nodules. These nodules showed a high signal intensity on T2-weighted magnetic resonance imaging. Subsequently, a diagnosis of PPGL was made based on elevated urinary excretion of adrenaline (355 µg/day [1937 nmol/day]; normal range: 3.4-26.9 µg/day; 18-146 nmol/day), noradrenaline (1690 µg/day [9989 nmol/day]; normal range: 48.6-168.4 µg/day; 287-995 nmol/day), and dopamine (53 000 µg/day [258 322 nmol/day]; normal range: 365-961.5 µg/day; 1779-4686 nmol/day). The 123I-metaiodobenzylguanidine scintigraphy and fluorodeoxyglucose positron emission tomography scan showed heterogenous uptake among the adrenal and the liver foci, respectively. Clustering analysis of previous PPGL cases highlighted the unique catecholamine profile of this case. These findings suggest a possibility that internodular heterogeneity between primary and metastatic foci on nuclear imaging may indicate varying differentiation grades and resultant catecholamine secretion. Further studies will be needed to verify these results and confirm this hypothesis.
Collapse
Affiliation(s)
- Keiko Yoshioka
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8641, Ishikawa, Japan
| | - Yujiro Nakano
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8641, Ishikawa, Japan
| | - Moeka Horichi
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8641, Ishikawa, Japan
| | - Daisuke Aono
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8641, Ishikawa, Japan
| | - Yumie Takeshita
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8641, Ishikawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8641, Ishikawa, Japan
| |
Collapse
|
48
|
Ying Y, Ju R, Wang J, Li W, Ji Y, Shi Z, Chen J, Chen M. Accuracy of machine learning in diagnosing microsatellite instability in gastric cancer: A systematic review and meta-analysis. Int J Med Inform 2025; 193:105685. [PMID: 39515046 DOI: 10.1016/j.ijmedinf.2024.105685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Significant challenges persist in the early identification of microsatellite instability (MSI) within current clinical practice. In recent years, with the growing utilization of machine learning (ML) in the diagnosis and management of gastric cancer (GC), numerous researchers have explored the effectiveness of ML methodologies in detecting MSI. Nevertheless, the predictive value of these approaches still lacks comprehensive evidence. Accordingly, this study was carried out to consolidate the accuracy of ML in the prompt detection of MSI in GC. METHODS PubMed, the Cochrane Library, the Web of Science, and Embase were retrieved up to March 20, 2024. The risk of bias in the encompassed studies was evaluated utilizing a risk assessment tool for predictive models. Models were then subjected to subgroup analysis based on the modeling variables. RESULTS A total of 12 studies, encompassing 11,912 patients with GC, satisfied the predefined inclusion criteria. ML models established in these studies were primarily based on pathological images, clinical features, and radiomics. The results suggested that in the validation sets, the pathological image-based models had a synthesized c-index of 0.86 [95 % CI (0.83-0.89)], with sensitivity and specificity being 0.86 [95 % CI (0.76-0.92)] and 0.83 [95 % CI (0.78-0.87)], respectively; radiomics feature-based models achieved respective values of 0.87 [95 % CI (0.81-0.92)], 0.77 [95 % CI (0.70-0.83)] and 0.81 [95 % CI (0.74-0.87)]; radiomics feature-based models + clinical feature-based models achieved respective values of 0.87 [95 % CI (0.81-0.93)], 0.78 [95 % CI (0.70-0.84)] and 0.79 [95 % CI (0.69-0.86)]. CONCLUSIONS ML has demonstrated optimal performance in detecting MSI in GC and could serve as a prospective early adjunctive detection tool for MSI in GC. Future research should contemplate minimally invasive or non-invasive, readily collectible, and efficient predictors to augment the predictive accuracy of ML.
Collapse
Affiliation(s)
- Yuou Ying
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Ruyi Ju
- Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jieyi Wang
- The Basic Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Wenkai Li
- Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Yuan Ji
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Zhenyu Shi
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jinhan Chen
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Mingxian Chen
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, Street Gucui No. 234, Region Xihu, Hangzhou 310012, Zhejiang Province, China.
| |
Collapse
|
49
|
Wang M, Guo X, Liu X, Huang L, Yang C. Artificial Intelligence-Guided Identification of IGFBP7 as a Critical Indicator in Lactic Metabolism Determines Immunotherapy Response in Stomach Adenocarcinoma. J Cell Mol Med 2025; 29:e70301. [PMID: 39788916 PMCID: PMC11717556 DOI: 10.1111/jcmm.70301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/28/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Due to considerable tumour heterogeneity, stomach adenocarcinoma (STAD) has a poor prognosis and varies in response to treatment, making it one of the main causes of cancer-related mortality globally. Recent data point to a significant role for metabolic reprogramming, namely dysregulated lactic acid metabolism, in the evolution of STAD and treatment resistance. This study used a series of artificial intelligence-related approaches to identify IGFBP7, a Schlafen family member, as a critical factor in determining the response to immunotherapy and lactic acid metabolism in STAD patients. Computational analyses revealed that a high lactic metabolism (LM) state was associated with poor survival in STAD patients. Further biological network-based investigations identified a key subnetwork closely linked to LM. Machine learning techniques, such as random forest and least absolute shrinkage and selection operator, highlighted IGFBP7 as a crucial indicator in STAD. Functional annotations showed that IGFBP7 expression was linked to important immune and inflammatory pathways. In vitro experiments demonstrated that silencing IGFBP7 suppressed cell proliferation and migration. Furthermore, heightened susceptibility to several chemotherapeutic drugs was linked to elevated IGFBP7 levels. In conclusion, this work sheds light on the mechanisms by which the lactate metabolism-related indicator IGFBP7 affects the tumour immune milieu and the response to immunotherapy in STAD. The results point to IGFBP7 as a possible therapeutic target and predictive biomarker for the treatment of STAD.
Collapse
Affiliation(s)
- Minghua Wang
- Department of General SurgeryThe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Xiaofei Guo
- Department of OncologyThe 962 Hospital of the Chinese People's Liberation Army Joint Logistic Support ForceHarbinHeilongjiangChina
| | - Xuyun Liu
- Department of General SurgeryThe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Lei Huang
- Department of General SurgeryThe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Chuang Yang
- Department of General SurgeryThe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| |
Collapse
|
50
|
Kim KT, Lee MH, Shin SJ, Cho I, Kuk JC, Yun J, Choi YY. Decorin as a key marker of desmoplastic cancer-associated fibroblasts mediating first-line immune checkpoint blockade resistance in metastatic gastric cancer. Gastric Cancer 2025; 28:12-26. [PMID: 39520589 DOI: 10.1007/s10120-024-01567-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Gastric cancer (GC) remains a significant cause of cancer-related mortality worldwide. Despite the transformative impact of immune checkpoint blockade (ICB) therapies across various cancers, only a minority of patients with metastatic GC (mGC) benefit, emphasizing the urgent need for precise biomarkers to predict therapeutic responses and optimize patient selection. METHODS In this multi-omics study, we conducted whole exome and transcriptome sequencing on 12 tumors from mGC patients treated with nivolumab as first-line therapy. To validate our findings, we performed whole transcriptome sequencing on 17 additional tumors and analyzed 45 tumors from public dataset (PRJEB25780) of patients who received ICB therapy as second or third-line treatment. Comprehensive multi-omics analyses were conducted using single-cell RNA sequencing (n = 5, GSE167297) and spatial transcriptome sequencing (n = 2, independent internal dataset). RESULTS ICB-sensitive tumors exhibited robust activation of the interferon response pathway, while ICB-resistant tumors displayed epithelial-mesenchymal transition signatures. Intriguingly, at the single-cell level, genes associated with ICB sensitivity were predominantly expressed in immune cells, whereas genes associated with resistance were primarily found in cancer-associated fibroblasts (CAFs), particularly the desmoplastic CAF (dCAF) subtype. We identified DCN as a hallmark dCAF marker, and high DCN expression was inversely correlated with PD-L1 levels, ICB resistance, and poor prognosis in mGC (log-rank p = 0.027). CONCLUSION This study elucidates the critical influence of the tumor microenvironment, specifically dCAFs, in mediating ICB resistance in mGC. Our findings highlight DCN as a representative marker for dCAF and a promising negative predictive biomarker for ICB response. These findings highlight the complex stromal-immune interactions and open avenues for personalized treatment for mGC.
Collapse
Affiliation(s)
- Ki Tae Kim
- Department of Molecular Genetics & Dental Pharmacology, School of Dentistry, Seoul National University, Seoul, Korea
- Dental Research Institute and Dental Multi-omics Center, Seoul National University, Seoul, South Korea
| | - Min Hee Lee
- Department of Radiology, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Su-Jin Shin
- Department of Pathology, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
| | - In Cho
- Department of Surgery, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Jung Cheol Kuk
- Department of Surgery, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Jina Yun
- Department of Medicine, Division of Hematology/Oncology, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Yoon Young Choi
- Department of Surgery, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|